You are on page 1of 13

NEUROL-1580; No.

of Pages 13

revue neurologique xxx (2015) xxx–xxx

Available online at

ScienceDirect
www.sciencedirect.com

Neuroepidemiology

Epidemiology of Parkinson’s disease

A. Elbaz a,b,c,*, L. Carcaillon d, S. Kab a,b,c, F. Moisan c


a
Inserm, centre for research in epidemiology and population health, U1018,
epidemiology of ageing and age related diseases, 94807 Villejuif, France
b
University Paris-Sud, UMRS 1018, 94807 Villejuif, France
c
Département santé travail, institut de veille sanitaire (InVS), 94415 Saint-Maurice, France
d
Département des maladies chroniques et des traumatismes, institut de veille sanitaire (InVS),
94415 Saint-Maurice, France

info article abstract

Article history: Parkinson’s disease (PD) is the second most common neurodegenerative disease after
Received 13 May 2015 Alzheimer’s. PD is considered a multifactorial disorder that results, in most cases, from
Received in revised form the combined effects of multiple risk and protective factors, including genetic and envi-
25 August 2015 ronmental ones. This review discusses some of the methodological challenges involved in
Accepted 1st September 2015 assessing the descriptive, prognostic and etiological epidemiological studies of PD, and
Available online xxx summarizes their main findings.
# 2015 Elsevier Masson SAS. All rights reserved.
Keywords:
Parkinson’s disease
Epidemiology

symptoms are most often unilateral or markedly asymme-


1. Parkinson’s disease trical; they become more pronounced as the disease pro-
gresses and eventually become a source of disability. A variety
Described by James Parkinson in 1817, Parkinson’s disease (PD) of non-motor symptoms can precede motor symptoms or
is a chronic neurodegenerative disease characterized by the develop over the course of the disease, and include consti-
loss of dopaminergic neurons in the substantia nigra which pation, anosmia, depression, psychosis, cognitive dysfunc-
leads to decreased levels of dopamine in the striatum and tion and dementia.
disrupted motor control. Neuronal eosinophilic inclusions Apart from a pathological examination of the brain, there is
known as Lewy bodies and aggregation of alpha-synuclein currently no other laboratory test or imaging technique that
protein are hallmarks of the disease. Many other neuronal cell can diagnose PD with certainty. Its diagnosis is purely clinical,
populations are also affected and account for the presence of and relies on medical history and neurological evaluation;
non-motor symptoms. The main pathophysiological mecha- additional tests may be useful for ruling out other causes of
nisms include mitochondrial dysfunction, abnormal aggrega- parkinsonism. Compared with the final diagnoses obtained at
tion of alpha-synuclein and oxidative stress [1]. autopsy, diagnoses made by neurologists are relatively good,
The cardinal signs of PD include rest tremor, slowness of albeit imperfect (positive predictive value = 76%) [2]; never-
movement, rigidity and postural instability. At disease onset, theless, they have improved over the past few decades [3]

* Corresponding author. Hôpital Paul-Brousse, bâtiment 15/16, 16, avenue Paul-Vaillant-Couturier, 94807 Villejuif cedex, France.
E-mail address: alexis.elbaz@inserm.fr (A. Elbaz).
http://dx.doi.org/10.1016/j.neurol.2015.09.012
0035-3787/# 2015 Elsevier Masson SAS. All rights reserved.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

2 revue neurologique xxx (2015) xxx–xxx

and can be further improved if movement-disorder specialists is likely to be more severe with older participants when
are involved (positive predictive value = 98%) [4]. exposure periods are distant in time and for detailed data. In
PD is the most common cause of parkinsonism. Other addition, PD may be responsible for cognitive impairment,
neurodegenerative diseases characterized by the presence of especially in those with more advanced disease, which can
parkinsonism, such as multiple system atrophy and pro- lead to recall biases.
gressive supranuclear palsy, are rare and their risk factors Increasingly, however, as a large number of studies are
have rarely been studied. The present review is restricted performed for specific questions, meta-analyses are becoming
solely to PD. available, thereby allowing for pooled analyses, examination
of sources of heterogeneity and testing for publication bias.

2. Epidemiological tools 2.3. Prognostic studies

2.1. Descriptive studies These are based on cohorts of PD patients followed over time.
The majority have been hospital-based or performed as part of
Door-to-door studies, where everyone in a given population is clinical trials, and rely on highly selected patients attending
screened and then examined to confirm the diagnosis, are referral centers (younger patients, familial forms of disease,
considered the reference approach. They have the advantage atypical presentations), leading to issues regarding data
of detecting undiagnosed patients. However, non-participa- generalizability. Many studies have included prevalent rather
tion may be a problem if it can influence the outcome, and the than incident patients, and there are as yet few studies
screening instrument needs to be highly sensitive. Also, these involving incident patients from the general population
studies are expensive and difficult to implement, and yet can followed over time [22]. More efforts are needed in this area,
usually detect only a limited number of affected persons. In and such studies should tend towards harmonization of
addition, different diagnostic criteria can have a major impact assessment tools.
on frequency estimates [5]. PD registries represent an
alternative [6], but their feasibility is not straightforward, as
there may be multiple sources of patients, the diagnosis is 3. Descriptive epidemiology
mainly clinical and there may be important variations in
access to specialized healthcare. 3.1. Incidence and prevalence
Death certificates fail to identify a large number of PD
patients because the disease is not always mentioned [7,8], PD incidence is usually comprised between 10 and 50/
and they can also lead to diagnostic misclassification. In a 100,000 person-years, and its prevalence between 100 and
recent Spanish study, PD was rarely reported as the primary 300/100,000 population [23,24]. Although it is the second most
cause of death (14.6%) [9]. Another study that searched for PD common neurodegenerative disease after Alzheimer’s, PD
mentioned anywhere on the death certificate reported a 57% remains relatively uncommon. However, due to the general
frequency [10]. aging of the population, the number of PD patients is expected
Databases of large drug claims are increasingly available to double by 2030 [25].
and offer an interesting opportunity to identify patients with PD frequency increases sharply with age. It is rare before
specific conditions. However, as antiparkinsonian drugs are age 50 years, and its incidence and prevalence both increase
not exclusively specific to PD, validation is an important step. progressively after age 60; based on a meta-analysis of
A few studies, including one in France, have shown that the prevalence studies, its prevalence rose from 107/100,000 per-
identification of PD patients using antiparkinsonian drug sons between ages 50 and 59 years to 1087/100,000 persons
claims is a valid approach, particularly when several drugs between 70 to 79 years [24]. PD is usually more common in
and dosages are taken into account [11–14]. men than women, with a male-to-female ratio of about
1.5 [26–28]. Possible explanations for this difference include
2.2. Etiological studies more frequent occupational exposures in men, neuropro-
tection from estrogens in women and X-linked genetic
Because the incidence of PD is not high, many epidemiological factors.
studies of the etiology of PD have relied on a case-control, In France, based on reimbursement data of antiparkinso-
rather than cohort, design. Only in a few large cohort studies nian drugs from the French National Health Insurance System
with long follow-up has PD diagnosis been ascertained and a (Système national d’information inter-régimes de l’assurance mala-
sufficient number of cases identified [15–21]. die), it is estimated that there were around 150,000 prevalent
While this review will not discuss in detail the advantages cases (prevalence = 2.30/1000 population) and 25,000 incident
and disadvantages of case-control vs cohort studies, it is cases (incidence = 0.39 per 1000 person-years) in 2010 (Fig. 1)
worth mentioning a few important points. In case-control [29].
studies based on prevalent cases, the collection of exposure Geographical differences in PD frequency have been
data is retrospective and involves subjects who already have reported, but are difficult to interpret because of differences
the disease. As PD mainly affects elderly people, this in finding cases and population characteristics. A collaborative
approach has obvious consequences for the assessment of study conducted in four European countries using similar
exposures over long periods of time, and the collected data case-finding methods and diagnostic criteria did not reveal
are at risk of being inaccurate or include missing values. This any differences [30]. On the other hand, a meta-analysis of six

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 3

Fig. 1 – Prevalence and incidence of Parkinson’s disease (PD) in France in 2010. The numbers of patients with prevalent and
incident PD were estimated from nationwide drug-claims databases based on social security systems [13,29]. The slight
decrease after age 85 years is partly explained by the lack of identification of patients in institutions with internal
pharmacies. The average age at PD onset was 74.8 years overall, and 74.0 years in men and 75.6 years in women.

studies found a lower prevalence in Africa than in either disease is progressive with worsening of the motor and non-
Europe or North America [31], whereas there was no motor symptoms responsible for the patient’s increasing
significant difference between African-Americans and Cau- disability. The neuropathological staging system proposed by
casians in the United States [32]. The prevalence and incidence Braak et al. [38] has allowed better characterization of the
of PD also appear to be slightly lower in Asian than in Western neuropathological basis underlying the progression of PD.
countries [24,33], although some studies have found similar Here, the focus is on three outcomes: death, dementia and
estimates [34]. It is unclear whether such differences are due cancer. Additional markers of clinical progression are dis-
to environmental factors or whether they reflect methodolo- cussed elsewhere [39].
gical differences across studies.
Very few studies have examined temporal trends in PD 3.2.1. Mortality
frequency. Diagnosis of the disease and rules for coding death Life expectancy and mortality risk in PD patients have been
certificates have evolved over time, which makes interpreting extensively studied [22]. The vast majority of studies found a
trends over time difficult [35]. A study in Minnesota, however, significantly higher risk of mortality in PD cases compared
has shown that the incidence of PD remained stable over a with people free of the disease of similar age and gender,
short period of time (1976–1990) [36], whereas a Finnish study although the magnitude of the risk varied across studies,
comparing the incidence and prevalence of PD in 1971 and ranging from 1.26 to 3.79 [22]. Some studies found no
1992 found increases only in men [37]. significant differences in survival related to PD [40–43], but
they lacked statistical power, had short follow-up durations or
3.2. Prognosis were based on selected populations. As shown in a meta-
analysis, differences in study design and, in particular,
Understanding PD prognosis is necessary for improving the inclusion of prevalent or incident cases and duration of
management and care of PD patients. Although there are follow-up can explain some of the heterogeneity [22]. Based on
effective symptomatic treatments, there is no cure, and the inception cohorts that followed incident PD cases, there was

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

4 revue neurologique xxx (2015) xxx–xxx

an estimated pooled mortality ratio of 1.52 (95% CI: 1.23–1.88) cancer showed a strong inverse association between smoking-
with little heterogeneity, a 5% annual decrease in survival of related cancers and PD [65]. However, a decreased risk of
PD patients and a mean median survival of 12.6 years from cancers unrelated to smoking (colon, rectum, prostate) has
disease onset compared with 16.0 years in non-PD subjects. also been shown, albeit weaker than for smoking-related
Mortality ratios increased with longer follow-ups. No robust cancers, thereby suggesting that other mechanisms may be
evidence was found that mortality decreased after the involved [65,66]. An alternative hypothesis involves genetic
introduction of levodopa. factors influencing cell-cycle regulation that may protect
There is remarkable individual variability in time to against cancer while predisposing to PD development; for
death after PD diagnosis, although individual prognostic instance, the parkin gene, one of the genes responsible for
factors are still poorly understood. When compared with recessive familial PD, plays a role in cancer etiology and
age-matched PD-free subjects, younger PD patients have a progression [67].
higher mortality risk than older patients [10,44,45]. The risk In contrast to the overall reduced risk of developing cancer,
of mortality increases with motor-function deterioration melanoma has been shown to occur with a greater frequency
[46], severity of axial impairment [47] and the presence in PD [68]. This association is found both before and after
of dementia [44,45,47]. As regards causes of death, the diagnosis, so levodopa treatment is unlikely to be an
leading ones in PD are heart disease, pneumonia and stroke explanation. Common genetic factors are also unlikely to
[40,48–51]. account for this association [66,69]. It has been suggested that
One study surveyed the records of 129 subjects with a shared embryonic origin of melanocytes and neurons in the
pathologically proven PD who were separated into five groups neural crest may be involved.
according to age at time of death to compare those who had In view of the complex relationship between cancer and PD,
reached the advanced stage of disease in terms of age. Four more research is needed to clarify the mechanisms underlying
milestones of advanced disease (frequent falls, visual hallu- these associations and the potential implications.
cinations, dementia and need for residential care) happened at
a similar time prior to death in each group, irrespective of the
age at which they occurred. These observations suggest that 4. Risk and protective factors
the disease can be subdivided into a variable early-middle
phase followed by the more stereotypical late period of PD is considered a multifactorial disorder that is caused, in
milestone accumulation, which appears to be less influenced most cases, by the combined effects of multiple factors,
by age at onset than the previous phase. The accumulation of including genetic and environmental ones. Approximately 10–
multiple milestones was associated with high scores for 15% of PD patients report a family history of PD among their
cortical Lewy bodies [52]. first-degree relatives [70]. In a minority of patients, a single
gene mutation associated with Mendelian transmission of the
3.2.2. Dementia disease is found; 15 causal genes are currently identified, and
Another major aspect of PD prognosis pertains to the their mutations are most often associated with a younger age
development of cognitive impairment and dementia. A of onset [71]. Genetic susceptibility has also been implicated in
systematic review reported that PD-related dementia repre- sporadic forms [72]. Although a genetic risk score, composed
sents 3–4% of all dementia patients [53]. Given the specificity of multiple variants independently associated with disease
and importance of PD-related dementia, specific diagnostic risk, has poor predictive power overall (area under the receiver
criteria have been developed for this outcome in PD [54,55]. operator curve = 0.616), it decreases with increasing age across
The annual incidence of dementia in PD varied from 30 to 95/ the continuum of age at onset, thereby suggesting that the
1000 person-years [39], and the relative risk of developing accumulation of common polygenic alleles with relatively low
dementia in PD was estimated to be between 1.7 and 5.9 effect sizes makes a greater contribution to early, rather than
compared with subjects free of PD [56–59]. The heterogeneity late, onset PD [73,74]. Similarly, twin studies have found low
across studies can be explained by differences in study design concordance for the disease except when the disease begins at
(including the source of cases), inclusion of prevalent or an early age [75,76]. Thus, in a large proportion of cases, PD is
incident cases, duration of follow-up, criteria used for not explained by genetic factors, especially when the disease
dementia and type of comparator group. The main factors does not appear in young patients, whereas epidemiological
influencing the risk of dementia in PD are PD duration [43,60], and toxicological studies provide important results for the role
older age at PD diagnosis [39,43,61,62] and greater PD severity of environmental factors.
[63,64]. A growing body of evidence suggests that dementia is It has become increasingly clear that PD starts many years
responsible for a considerable amount of the reduced life before the motor symptoms become evident and PD is
expectancy in PD patients [47,57,60]. diagnosed. Brain imaging techniques show that motor signs
appear when dopaminergic neuronal loss is already signifi-
3.2.3. Cancer cant. Cohort studies show that non-motor symptoms, such as
A large body of epidemiological evidence shows a decreased anosmia, constipation or rapid eye movement (REM) sleep
cancer risk in PD. A meta-analysis of 29 studies found an disorders, may be present for years before the onset of motor
inverse relationship between cancer and PD (OR: 0.73, 95% CI: signs – up to 20 years in some studies [77]. These findings may
0.63–0.83) [65]. PD patients have lower lifelong tobacco use help to define more precisely the periods of vulnerability to
than non-PD subjects (see below), and smoking is a major risk better assess relevant exposures; it is also possible that the
factor for many cancers; studies looking at different types of effects of repeated insults may accumulate before motor

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 5

symptoms appear, in which case, assessing cumulative in two case-control studies [87,88]. A cohort study of more
exposures before the onset of motor symptoms is warran- than 6800 people followed for 40 years could find no
ted. Nevertheless, the fact that motor and non-motor association between sensation-seeking and PD [89]. Other
symptoms are present before diagnosis needs to be traits, such as being introverted, conservative, socially
carefully considered in the interpretation of epidemiologi- vigilant, tense [90] or anxious, have been found to be
cal findings, as they may influence exposures and induce associated with PD [91]. However, an inverse association
associations (reverse causation). between smoking and PD has been reported as early as
Many exposures have been examined in relation to PD, and 20 years before disease onset [80,81]; this hypothesis would
this review addresses some of the most commonly examined therefore be plausible if there was a very long prodromal
ones. Where meta-analyses are available, these sources have phase. Third, a shared smoking and PD risk factor could be
been favored, with descriptions of specific studies that bring hypothesized, but it is unlikely to be genetic, as the inverse
additional information. The interested reader can access a association between smoking and PD is also present when
more detailed review that thoroughly examines risk and comparing affected twins with their healthy, non-PD, co-twins
protective factors elsewhere [78]. [92,93].
In recent years, investigators have devised novel study
4.1. Smoking designs to address these issues. Two case-control studies
focused on passive smoking, which is less likely to be affected
For over 30 years, numerous studies have reported an inverse by genetic factors, personality traits or other behaviors, and
association between smoking and PD. A meta-analysis of found an inverse association between PD and living or working
44 case-control and four cohort studies found a 60% lower PD with smokers [94,95]. Another case-control study evaluated
risk among ever-smokers compared with never-smokers [79]. the dietary intake of nicotine derived from plants in the
Despite different methodologies and populations, between- Nicotiana species of the Solanaceae family (tomatoes, peppers,
study heterogeneity was low to moderate. The same rela- eggplant, potatoes) and found an inverse association [96].
tionship was present in an analysis of individual data from On the other hand, the association may reflect biological
eight case-control studies and thee cohort studies from the US mechanisms that remain poorly elucidated. One study
[80]. This robust inverse association is characterized by a dose– showed inhibition of the monoamine oxidase B (MAO-B)
response relationship, with decreasing associations as the enzyme in the brains of smokers, and suggested that smoking
number of pack-years or years of smoking increase [79]. More may have a protective effect for dopaminergic neurons [97].
recently, a cohort study sought to distinguish the respective Others have suggested that nicotine may have antioxidant
roles of duration and intensity of smoking, and found that properties [98]. A recent study showed that, of five compounds
duration seemed to be more important than intensity [81]. found in cigarette smoke (anabasine, cotinine, hydroquinone,
This observation is important as it suggests that many years of nicotine and nornicotine), nicotine and hydroquinone led to
smoking are required before observing a decreased risk of PD, the greatest inhibition of alpha-synuclein fibrillization [99].
which represents a significant limitation for testing any One of the main difficulties in toxicological studies is the large
potential therapeutics. number of smoke components (over 4000 different molecules);
The inverse relationship between smoking and PD is seen nevertheless, more work needs to be done in this area to better
in both men and women [80,81], whereas no interaction understand this intriguing association and to elucidate
between smoking and education or exposure to pesticides has whether it is causal or not.
been demonstrated [80,82]. A similar relationship was obser-
ved with other forms of tobacco use: PD patients less often 4.2. Coffee- and tea-drinking
smoke pipes or cigars and chew tobacco less often than do
controls [80]. In addition, a stronger inverse association Caffeine and some of its metabolites (theophylline, paraxan-
between smoking and PD was observed among cases with thine) are antagonists of adenosine A2A receptors, and in-vivo
younger onset of disease (< 75 years) compared with cases studies in mice have demonstrated their ability to attenuate
with older onset of disease ( 75 years) [80]. On the other hand, the neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahy-
cohort studies of PD patients suggest that smoking has no dropyridine (MPTP) [100].
effect on disease progression [83]. A meta-analysis (eight case-control and four cohort
The causality of the association remains controversial, and studies) of the relationship between coffee-drinking and PD
several alternative hypotheses have been proposed [84,85]. found a 30% decreased risk for coffee drinkers compared with
First, survival bias does not appear to be responsible because non-drinkers [79]. This relationship was independent of
several cohort studies have confirmed it; in addition, smoking smoking, and risk reduction increased as the number of cups
does not appear to be associated with higher mortality among per day increased. Also, a recent cohort study reported that the
cases vs non-cases [10]. Second, reverse causation may play a risk of PD decreased by 40% among those who drank more
role: PD patients may stop smoking more often or begin than three cups of coffee per day [101].
smoking less often before disease onset than those who do not Other studies have assessed the total intake of caffeine
develop PD [86] due to a reduced appetite for cigarettes or less based on coffee, tea, cola and chocolate consumption. A meta-
responsiveness to nicotine during the prodromal phase of PD. analysis of 26 studies reported a 25% reduction in PD risk
It has also been hypothesized that prodromal personality among caffeine consumers with a dose–response relationship
traits associated with a decreased appetite for smoking could [102]. These results are not explained by smoking. Further-
play a role. Low novelty-seeking has been associated with PD more, the association between caffeine intake and PD is

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

6 revue neurologique xxx (2015) xxx–xxx

observed in people who do not drink coffee, and whose working in other types of agriculture [110]. Among specific
sources of caffeine are mainly tea and colas [103]. pesticides, there is some evidence of an association for
Some studies have separately assessed coffee with and pesticides that induce oxidative stress or inhibit mitochon-
without caffeine, and no decreased risk of PD was found with drial complex I [111] and, in particular, organochlorine
decaffeinated coffee [15]. Similarly, one study found an insecticides [18,112].
association between black tea (which contains caffeine) and In addition to professional exposures, studies performed in
PD, but not with green tea (which does not contain caffeine) California using a pesticide registry and geographical infor-
[20]. Thus, caffeine seems to be the best candidate to explain mation systems found an increased risk of PD for people
this association. exposed in their residential environment [113–115].
The relationship between caffeine or coffee and PD appears Toxicological evidence of neurotoxic effects of some
to be less strong in women than in men [102]. An interaction pesticides is reviewed elsewhere [116–118]. Given the evi-
between coffee and postmenopausal hormone replacement dence, since 2012 in France, PD has been recognized as an
therapy (HRT) has been suggested to explain this difference occupational disease for agricultural workers who fulfil
[104], as the inverse association was observed only in women specific conditions: diagnosis confirmed by a neurologist;
not taking HRT. and professional exposure to pesticides for 10 or more years.
The same types of biases as cited for smoking could play a
role here, but have been less often studied with coffee and 4.4. Brain injury
caffeine. However, adenosine A2A receptors are abundantly
expressed on striatal neurons and, in animal models, their In a recent meta-analysis of 22 studies (only one cohort study),
blockade extends the efficacy of levodopa. This suggests that a history of head injury with loss of consciousness was
adenosine A2A receptor antagonists may be useful in PD associated with an increased risk of developing PD (OR: 1.57,
patients with motor fluctuations. However, while the results 95% CI: 1.35–1.83) [119]. This association also persisted in an
were inconclusive for the first drugs tested in humans analysis restricted to 13 studies of higher quality (OR: 1.46, 95%
(istradefylline, preladenant), additional clinical trials are CI: 1.33–1.82). There was, however, some evidence in favor of
underway to test the safety and efficacy of other publication bias that may have led to overestimation of the
A2A antagonists (tozadenant) – with encouraging results so association.
far [105,106]. In a case-control study based on twin pairs discordant for
PD, a dose–effect relationship was observed based on the
4.3. Pesticides number of head injuries, with more severe head injuries
associated with a greater risk of PD [120]. This is in agreement
The hypothesis of a link between PD and pesticide exposure with other studies showing stronger associations for injuries
appeared in the early 1980s, after several cases of parkinso- resulting in unconsciousness or hospitalization than for
nism followed intravenous injection of MPTP. MPTP is milder traumas [121,122].
metabolized into 1-methyl-4-phenylpyridinium (MPP+), an Several possible mechanisms might explain this associa-
inhibitor of the mitochondrial respiratory chain with neuro- tion. Head trauma and its repair process trigger inflammatory
toxic properties in dopaminergic cells. The molecule has a mechanisms that may be deleterious to the brain. Head
chemical structure similar to that of paraquat, a non-selective trauma can disrupt the blood–brain barrier, allowing neuro-
herbicide marketed since the 1960s and widely used ever toxins to penetrate the brain [122,123]. Head trauma may also
since. Following this observation, numerous studies have trigger aggregation of alpha-synuclein protein; one study
examined the relationship of farming and exposure to showed that the association between head trauma and PD was
pesticides with PD. stronger among people who carry a genetic polymorphism
A meta-analysis of 46 studies reported that the risk of PD is associated with higher levels of alpha-synuclein, thus raising
about 1.6 times higher in people exposed to pesticides, with a the hypothesis that head injury may initiate and/or accelerate
stronger association for herbicides (OR: 1.40) and insecticides neurodegeneration when levels of synuclein are high [124].
(OR: 1.50), albeit with significant heterogeneity across studies. There are, however, possible sources of bias that have been
The association was weaker for fungicides (OR: 1.0), but these addressed in more recent studies. Recall bias may be an issue,
compounds were examined in a limited number of studies as PD patients may be prone to overreporting previous head
[107]. Two other meta-analyses found similar results [108,109]. traumas as a possible cause of disease. However, a few studies
Exposure assessment represents the main limitation of have relied on medical records and found similar associations
several studies: few studies collected information on the types [122,123]; thus, recall bias is unlikely to fully explain the
of products, many combined professional and domestic association. Reverse causation may be another issue, as
exposures, few performed gender-stratified analyses, and patients may be at higher risk of falls during the prodromal
very few assessed dose–effect relationships or involved work- phase, when subtle motor signs are already present. In a large
exposure matrices or industrial hygienists [107]. A recent Swedish registry that assessed head injuries based on medical
French study performed among farmers reported a stronger records, head injuries with hospitalization were associated
dose–effect relationship for intensity of exposure than for with PD [123]. The authors excluded head traumas that
duration [110]. The study also showed that the association occurred at 1, 5, 10 and > 10 years before the diagnosis;
with pesticides was present for tremor-dominant PD, the most although the odds ratio (OR) was markedly attenuated, there
typical form of PD. In addition, there was a stronger was still a significant 20% increased risk for head injuries
association for farmers working in vineyards than for those occurring > 10 years before diagnosis [123]. Furthermore, in a

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 7

study from Canada, associations between PD and head trauma up in their sensitivity analyses, found consistent results, but
were not substantially attenuated by excluding head traumas the follow-up may not have been long enough and was thus
that occurred at 5, 10 and 20 years before PD diagnosis [121]. insufficient to fully exclude reverse causation as a potential
explanation.
4.5. Solvents Additional studies have examined the role of UA in relation
to disease progression and linked higher UA concentrations to
Solvents represent a broad class of compounds with many less severe progression [130], but not with risk of dementia
commercial applications, including metal degreasing, dry [131]. One study found that the risk of progression to disability
cleaning, paint thinners and detergents. Therefore, exposure requiring dopaminergic treatment increased with an increas-
is relatively widespread at workplaces and at home. Many ing number of SLC2A9 alleles associated with lower UA
solvents are lipophilic and able to enter the central and concentrations [132]. The Safety of Urate Elevation in PD
peripheral nervous systems [125]. (SURE-PD) study, a randomized, double-blind, placebo-
Despite differences in chemical structure, especially controlled, dose-ranging trial of inosine, is currently under-
between chlorinated and non-chlorinated agents, solvents way and has reported that inosine is generally safe, tolerable
are treated as a single entity in the majority of studies. In a and effective in raising serum and cerebrospinal fluid levels of
meta-analysis of 16 case-control studies, PD was associated UA in early PD [133].
with solvents (OR: 1.35, 95% CI: 1.09–1.67) [109]. The associa-
tion was slightly stronger when only six studies of the highest 4.7. Consumption of dairy products
quality were considered (OR: 1.58, 95% CI: 1.23–2.04). However,
no association was reported in two cohort studies that Higher consumption of dairy products has been associated
collected data on solvents [16,126]. with increased PD risk in several cohort studies; in a meta-
Specific solvents have been investigated in greater detail in analysis of four such studies, the combined relative risk for the
a twin study [127]. Lifetime exposures to six solvents was highest intake was 1.6 (95% CI: 1.3–2.0) [134]. An association
inferred by industrial hygienists and/or occupational medi- was also reported in a recent prospective cohort from Greece
cine physicians who were all unaware of PD status, but used (OR: 1.33, 95% CI: 1.07–1.65) after adjusting for several
work-related task-specific structured questionnaires, exten- confounders, including gender, smoking, farming, caffeine
sive probability databases and multiple reference sources. and energy intakes [135]; this association was independent of
Based on 99 discordant twin pairs (49 monozygotic, 50 mono- calcium intake, as no association was observed with either
zygotic), the smoking-adjusted PD risk was increased for those cheese or yoghurt.
who had ever been exposed to trichloroethylene (OR: 6.1, 95% Potential explanations for this association include conta-
CI: 1.2–33) and perchloroethylene (OR: 10.5, 95% CI: 0.97–113.0). mination of dairy products with neurotoxins or decreased
This finding is consistent with a rodent model of trichloroe- circulating levels of UA associated with greater dairy food
thylene-induced parkinsonism, which revealed key patholo- consumption [134]. Again, reverse causation cannot be
gical and neurochemical PD features [128]. Further studies of excluded.
the association between PD and solvents are needed to better
understand which products are more likely to be involved, to 4.8. Anti-inflammatory drugs
characterize any dose–effect relationships and to identify the
relevant windows of exposure. Because neuroinflammatory factors are believed to play a role
in the pathogenesis of PD [136,137] and non-steroidal anti-
4.6. Uric acid inflammatory drugs (NSAIDs) exhibit neuroprotective effects
in animal PD models, some studies have investigated whether
Serum uric acid (UA) plays a major role as an antioxidant, a anti-inflammatory medications, especially NSAIDs, confer
property that may explain why UA confers protection in protection against PD [138].
cellular PD models and attenuates toxin-induced loss of In a meta-analysis of seven studies (five case-control and
primary neurons in lab cultures [129]. two cohort studies), a modest reduction in PD risk was
Several epidemiological studies have examined the asso- associated with NSAIDs (OR: 0.87, 95% CI: 0.75–1.00) [139]. An
ciation between UA or gout and PD, and most reported an inverse association was found for non-aspirin NSAIDs (OR:
inverse association with a dose–effect relationship in some. In 0.85, 95% CI: 0.77–0.94), including ibuprofen (OR: 0.75, 95% CI:
a meta-analysis of cohort and nested case-control studies in 0.64–0.89), but not for aspirin (OR: 1.08, 95% CI: 0.92–1.27). For
which UA was measured before diagnosis, high levels non-aspirin NSAIDs, analyses stratified by duration of use and
(> 6.8 mg/dL) were associated with a 33% risk reduction (OR: intensity of use yielded results consistent with a dose–
0.67, 95% CI: 0.50–0.91) [130]. This association was present in response relationship. Ibuprofen activates peroxisome pro-
men, but not in women [130], a difference that could be due to liferator-activated receptor (PPAR)-g, and PPAR agonists have
lower UA levels or increased mitochondrial respiration been shown to exert neuroprotective actions against oxidative
efficiency related to estrogens in women. damage, inflammation and apoptosis in several neurodege-
Although this inverse association may be causal, it could nerative disorders, including PD, both in vivo and in vitro [140].
also be due to reverse causation, as PD patients may change These findings support the hypothesis that non-aspirin
their diet or other behaviors years before the diagnosis NSAIDs may confer protection against PD and warrant further
because of constipation or other non-motor symptoms. Some investigation as they may have important clinical implica-
studies, which excluded cases during the first years of follow- tions. Nevertheless, potential biases may account for these

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

8 revue neurologique xxx (2015) xxx–xxx

findings [139]. First, NSAID use for non-motor manifestations (CCB) use was associated with a 25% reduction in PD risk; this
several years before PD diagnosis, such as pain-related pattern was evident both for dihydropyridine and non-
stiffness, can happen. Second, the lower mortality risk in dihydropyridine CCBs, albeit based on only two studies
PD patients who use NSAIDs compared with other PD patients [148]. Another study followed hypertensive Chinese patients
may also play a role. who were free of PD, dementia and stroke, and examined the
association between different antihypertensive drugs and
4.9. Vascular risk factors, statins and calcium-channel incidence of PD [149]. The use of dihydropyridine, but not non-
blockers dihydropyridine, CCBs, and of centrally acting CCBs rather
than peripheral ones, were associated with a reduced risk of
The role of vascular risk factors has been investigated PD. In addition to observational studies, a phase-II randomi-
primarily because of their role in other neurodegenerative zed, double-blind, parallel-group trial – Safety, tolerability and
diseases, such as Alzheimer’s. In addition, some of the drugs efficacy assessment of dynacirc CR in Parkinson disease
used to treat vascular risk factors have been shown to exert (STEADY-PD) – included 99 subjects with early PD not
neuroprotective effects. requiring dopaminergic therapy randomized to either 5, 10
The evidence regarding diabetes is inconclusive, as studies or 20 mg of isradipine CR (a dihydropyridine CCB shown to be
have reported conflicting results [78]. One prospective study neuroprotective in animal models of PD) or a matching
found a modest increased risk of PD associated with diabetes; placebo daily [150]. Tolerability of isradipine was dose-
however, the diagnosis of diabetes was clustered around the PD dependent, and the trial established 10 mg as the maximum
diagnosis, and was more apparent among men with short tolerable dosage of those tested. However, while there were no
diabetes duration and those without diabetes complications statistically significant differences in efficacy between the
[48]. Therefore, this association may be explained by surveil- active treatments or placebo, there was a suggestion of a trend
lance bias, and additional well-designed studies are necessary. towards increasing benefit with the 10-mg and 20-mg dosages.
Studies of hypertension show no association or lower risk A large placebo-controlled trial is now required to assess the
of PD in hypertensive subjects [78], a finding that may be efficacy of isradipine 10 mg/day in PD.
explained by reverse causation when blood pressure is
measured at around the time of PD diagnosis. Thus, additional 4.10. Gene–environment interactions
studies with midlife assessment of hypertension are needed.
Regarding cholesterol, a few prospective studies have In recent years, there has been a growing interest in
reported an inverse association between increasing levels of investigating gene–environment interactions in PD. One study
cholesterol and PD [141–143]. Secondary analysis of the performed in France among male farmers heavily exposed to
DATATOP trial has also provided preliminary evidence that pesticides found that exposures to organochlorine insecticides
higher total serum cholesterol concentrations may be asso- interact with the ABCB1 gene (which codes for P-glycoprotein, a
ciated with modest slowing of the clinical progression of PD transmembrane transporter acting as an active efflux pump for
[144]. As for other biomarkers, this inverse association may be a wide range of endogenous molecules and xenobiotics at the
causal, but it may also be due to reverse causation as PD blood–brain barrier) to increase PD risk. Thus, in men
patients may change their diet or other behaviors years before professionally exposed to organochlorines, polymorphisms
the diagnosis. associated with a reduced ability of ABCB1 to clear xenobiotics
Statins are widely used worldwide and are effective for from the brain increased the risk of PD [151]. These findings
reducing cholesterol levels. One meta-analysis of eight (five support the hypothesis of gene-by-pesticides interactions in
case-control and three cohort) studies showed a 20% risk the development of PD, and need to be replicated in larger
reduction of PD in statin users; there was, however, marked studies and other populations. There are other examples of
heterogeneity across studies and evidence of publication bias potential interactions between pesticides and other genes
[145]; further studies, and particularly of the role of long-term [152–155], or head trauma and the alpha-synuclein gene [124].
statin use, are needed. One recent study evaluated the These studies pose a number of methodological challenges,
association between discontinuation of statin use and the including the need for appropriate exposure-assessment
incidence of PD in a Chinese population free of PD that had methods and very large sample sizes, as the number of
initiated statin therapy [146]. Continued use of lipophilic subjects needed to detect interactions is usually large.
statins was associated with a 60% decreased risk of PD Replication is another major issue, as few of the above-
compared with statin discontinuation; there was no associa- mentioned examples have been reproduced to date. One
tion between hydrophilic statins and PD incidence. It has been report using a genome-wide approach to investigate genes
hypothesized that the beneficial role of statins in PD may be interacting with coffee-drinking showed evidence of an
the result of attenuation of neuroinflammatory processes due interaction with the GRIN2A gene [156]. Yet, upon closer
to inhibition of the expression of inflammatory mediators, inspection of the data, the interaction was explained by an
such as interleukin-6 and tumor necrosis factor-a, and association between coffee-drinking and the gene, but in the
protection against reactive oxygen species. controls and not among the cases. A further large-scale
The basal activity of dopaminergic neurons is driven by replication effort failed to reproduce the finding and found no
voltage-dependent L-type Ca2+ channels, and blocking Cav1.3 association among controls either [157].
Ca2+ channels in adult neurons protects them in models of PD Epigenetics is another emerging field that may represent a
both in vitro and in vivo [147]. A meta-analysis of five potential mechanism to account for the effects of environ-
observational studies showed that calcium-channel blocker mental factors in PD [158,159]. Epidemiological studies where

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 9

biological samples have been obtained have the potential to [9] Benito-Leon J, Louis ED, Villarejo-Galende A, Romero JP,
assess epigenetic changes in PD-related and other genes, and Bermejo-Pareja F. Under-reporting of Parkinson’s disease
on death certificates: a population-based study (NEDICES).
the influence of environmental factors on these changes. One
J Neurol Sci 2014;347(1–2):188–92.
issue, however, is to what extent epigenetic changes seen in
[10] Elbaz A, Bower JH, Peterson BJ, Maraganore DM,
peripheral tissues can be correlated with epigenetic changes McDonnell SK, Ahlskog JE, et al. Survival study of
in the brain. Parkinson disease in Olmsted County, Minnesota. Arch
Neurol 2003;60(1):91–6.
[11] Chio A, Magnani C, Schiffer D. Prevalence of Parkinson’s
5. Conclusion disease in Northwestern Italy: comparison of tracer
methodology and clinical ascertainment of cases. Mov
Disord 1998;13(3):400–5.
The identification of several PD genes has provided important [12] van de Vijver DA, Stricker BH, Breteler MM, Roos RA,
clues towards understanding the pathophysiology of the Porsius AJ, de Boer A. Evaluation of antiparkinsonian
disease. However, the proportion of cases explained by these drugs in pharmacy records as a marker for Parkinson’s
mutations is small and, in most cases, PD is likely to result disease. Pharm World Sci 2001;23(4):148–52.
from the combined effects of several factors with weak-to- [13] Moisan F, Gourlet V, Mazurie JL, Dupupet JL, Houssinot J,
Goldberg M, et al. Prediction model of Parkinson’s disease
moderate effects.
based on antiparkinsonian drug claims. Am J Epidemiol
Through different approaches, epidemiological studies
2011;174(3):354–63.
have provided important clues towards a better understand- [14] Chillag-Talmor O, Giladi N, Linn S, Gurevich T, El-Ad B,
ing of PD progression and prognosis, as well as evidence in Silverman B, et al. Estimation of Parkinson’s disease
favor of the role of environmental exposures in disease survival in Israeli men and women, using health
etiology. The degree of evidence is variable, although some maintenance organization pharmacy data in a unique
may have important clinical implications and are currently approach. J Neurol 2013;260(1):62–70.
[15] Ascherio A, Zhang SM, Hernan MA, Kawachi I, Colditz GA,
being explored through clinical trials. Epidemiological studies
Speizer FE, et al. Prospective study of caffeine
have generated results that can be studied experimentally to
consumption and risk of Parkinson’s disease in men and
understand the mechanisms leading from such exposures to women. Ann Neurol 2001;50(1):56–63.
neuronal loss. Thus, multidisciplinary teamwork involving [16] Ascherio A, Chen H, Weisskopf MG, O’Reilly EJ,
epidemiologists, toxicologists and geneticists is essential to McCullough ML, Calle EE, et al. Pesticide exposure and risk
better understand the etiology of PD. for Parkinson’s disease. Ann Neurol 2006;60(2):197–203.
[17] Chen H, Mosley TH, Alonso A, Huang X. Plasma urate and
Parkinson’s disease in the Atherosclerosis Risk in
Disclosure of interest Communities (ARIC) study. Am J Epidemiol
2009;169(9):1064–9.
[18] Weisskopf MG, Knekt P, O’Reilly EJ, Lyytinen J, Reunanen
The authors declare that they have no competing interest. A, Laden F, et al. Persistent organochlorine pesticides in
serum and risk of Parkinson disease. Neurology
2010;74(13):1055–61.
references [19] de Rijk MC, Breteler MM, Graveland GA, Ott A, Grobbee DE,
van der Meche FG, et al. Prevalence of Parkinson’s disease
in the elderly: the Rotterdam Study. Neurology
[1] Obeso JA, Rodriguez-Oroz MC, Goetz CG, Marin C, 1995;45(12):2143–6.
Kordower JH, Rodriguez M, et al. Missing pieces in the [20] Tan LC, Koh WP, Yuan JM, Wang R, Au WL, Tan JH, et al.
Parkinson’s disease puzzle. Nat Med 2010;16(6):653–61. Differential effects of black versus green tea on risk of
[2] Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of Parkinson’s disease in the Singapore Chinese Health
clinical diagnosis of idiopathic Parkinson’s disease: a Study. Am J Epidemiol 2008;167(5):553–60.
clinicopathological study of 100 cases. J Neurol Neurosurg [21] Petrovitch H, Ross GW, Abbott RD, Sanderson WT, Sharp
Psychiatry 1992;55(3):181–4. DS, Tanner CM, et al. Plantation work and risk of
[3] Horvath J, Burkhard PR, Bouras C, Kovari E. Etiologies of Parkinson disease in a population-based longitudinal
Parkinsonism in a century-long autopsy-based cohort. study. Arch Neurol 2002;59(11):1787–92.
Brain Pathol 2013;23(1):28–33. [22] Macleod AD, Taylor KS, Counsell CE. Mortality in
[4] Hughes AJ, Daniel SE, BenShlomo Y, Lees AJ. The accuracy Parkinson’s disease: a systematic review and meta-
of diagnosis of parkinsonian syndromes in a specialist analysis. Mov Disord 2014;29(13):1615–22.
movement disorder service. Brain 2002;125(4):861–70. [23] Von Campenhausen S, Bornschein B, Wick R, Botzel K,
[5] de Rijk MC, Rocca WA, Anderson DW, Melcon MO, Breteler Sampaio C, Poewe W, et al. Prevalence and incidence of
MM, Maraganore DM. A population perspective on Parkinson’s disease in Europe. Eur
diagnostic criteria for Parkinson’s disease. Neurology Neuropsychopharmacol 2005;15(4):473–90.
1997;48(5):1277–81. [24] Pringsheim T, Jette N, Frolkis A, Steeves TD. The
[6] Strickland D, Bertoni JM. Parkinson’s prevalence estimated prevalence of Parkinson’s disease: a systematic review
by a state registry. Mov Disord 2004;19(3):318–23. and meta-analysis. Mov Disord 2014;29(13):1583–90.
[7] Phillips NJ, Reay J, Martyn CN. Validity of mortality data for [25] Dorsey ER, Constantinescu R, Thompson JP, Biglan KM,
Parkinson’s disease. J Epidemiol Community Health Holloway RG, Kieburtz K, et al. Projected number of people
1999;53(9):587–8. with Parkinson disease in the most populous nations, 2005
[8] Beyer MK, Herlofson K, Arsland D, Larsen JP. Causes of through 2030. Neurology 2007;68(5):384–6.
death in a community-based study of Parkinson’s disease. [26] Wooten GF, Currie LJ, Bovbjerg VE, Lee JK, Patrie J. Are men
Acta Neurol Scand 2001;103(1):7–11. at greater risk for Parkinson’s disease than women? J
Neurol Neurosurg Psychiatry 2004;75(4):637–9.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

10 revue neurologique xxx (2015) xxx–xxx

[27] Elbaz A, Bower JH, Maraganore DM, McDonnell SK, [45] Posada IJ, Benito-Leon J, Louis ED, Trincado R, Villarejo A,
Peterson BJ, Ahlskog JE, et al. Risk tables for parkinsonism Medrano MJ, et al. Mortality from Parkinson’s disease: a
and Parkinson’s disease. J Clin Epidemiol 2002;55(1):25–31. population-based prospective study (NEDICES). Mov
[28] Taylor KS, Cook JA, Counsell CE. Heterogeneity in male to Disord 2011;26(14):2522–9.
female risk for Parkinson’s disease. J Neurol Neurosurg [46] Forsaa EB, Larsen JP, Wentzel-Larsen T, Alves G. What
Psychiatry 2007;78(8):905–6. predicts mortality in Parkinson disease? A prospective
[29] Direction de la recherche dédleds. Etat de santé de la population-based long-term study. Neurology
population en France en 2015. Ministère des Affaires 2010;75(14):1270–6.
sociales, de la Santé et des Droits des femmes 2015. [47] Levy G, Tang MX, Louis ED, Cote LJ, Alfaro B, Mejia H, et al.
[30] de Rijk MC, Tzourio C, Breteler MMB, Dartigues JF, The association of incident dementia with mortality in PD.
Amaducci L, LopezPousa S, et al. Prevalence of Neurology 2002;59(11):1708–13.
parkinsonism and Parkinson’s disease in Europe: the [48] Driver JA, Smith A, Buring JE, Gaziano JM, Kurth T,
EUROPARKINSON collaborative study. J Neurol Neurosurg Logroscino G. Prospective cohort study of type 2 diabetes
Psychiatry 1997;62(1):10–5. and the risk of Parkinson’s disease. Diabetes Care
[31] Okubadejo NU, Bower JH, Rocca WA, Maraganore DM. 2008;31(10):2003–5.
Parkinson’s disease in Africa: a systematic review of [49] Duarte J, Garcia Olmos LM, Mendoza A, Claveria LE. The
epidemiologic and genetic studies. Mov Disord natural history of Parkinson’s disease in the province of
2006;21(12):2150–6. Segovia: mortality in a longitudinal study (20-year follow-
[32] Schoenberg BS, Osuntokun BO, Adeuja AO, Bademosi O, up). Acta Neurol Scand 2013;127(5):298–300.
Nottidge V, Anderson DW, et al. Comparison of the [50] Pennington S, Snell K, Lee M, Walker R. The cause of death
prevalence of Parkinson’s disease in black populations in in idiopathic Parkinson’s disease. Parkinsonism Relat
the rural United States and in rural Nigeria: door-to-door Disord 2010;16(7):434–7.
community studies. Neurology 1988;38(4):645–6. [51] Willis AW, Schootman M, Kung N, Evanoff BA, Perlmutter
[33] Muangpaisan W, Hori H, Brayne C. Systematic review of JS, Racette BA. Predictors of survival in patients with
the prevalence and incidence of Parkinson’s disease in Parkinson disease. Arch Neurol 2012;69(5):601–7.
Asia. J Epidemiol 2009;19(6):281–93. [52] Kempster PA, O’Sullivan SS, Holton JL, Revesz T, Lees AJ.
[34] Zhang ZX, Anderson DW, Huang JB, Li H, Hong X, Wei J, Relationships between age and late progression of
et al. Prevalence of Parkinson’s disease and related Parkinson’s disease: a clinicopathological study. Brain
disorders in the elderly population of greater Beijing, 2010;133(Pt 6):1755–62.
China. Mov Disord 2003;18(7):764–72. [53] Aarsland D, Zaccai J, Brayne C. A systematic review of
[35] Goldacre MJ, Duncan M, Griffith M, Turner MR. Trends in prevalence studies of dementia in Parkinson’s disease.
death certification for multiple sclerosis, motor neuron Mov Disord 2005;20(10):1255–63.
disease, Parkinson’s disease and epilepsy in English [54] Emre M, Aarsland D, Brown R, Burn DJ, Duyckaerts C,
populations 1979-2006. J Neurol 2010;257(5):706–15. Mizuno Y, et al. Clinical diagnostic criteria for dementia
[36] Rocca WA, Bower JH, McDonnell SK, Peterson BJ, associated with Parkinson’s disease. Mov Disord
Maraganore DM. Time trends in the incidence of 2007;22(12):1689–707.
parkinsonism in Olmsted County, Minnesota. Neurology [55] Dubois B, Burn D, Goetz C, Aarsland D, Brown RG, Broe GA,
2001;57(3):462–7. et al. Diagnostic procedures for Parkinson’s disease
[37] Kuopio AM, Marttila RJ, Helenius H, Rinne UK. Changing dementia: recommendations from the movement disorder
epidemiology of Parkinson’s disease in southwestern society task force. Mov Disord 2007;22(16):2314–24.
Finland. Neurology 1999;52(2):302–8. [56] Aarsland D, Andersen K, Larsen JP, Lolk A, Kragh-Sorensen
[38] Braak H, Del TK, Rub U, de Vos RA, Jansen Steur EN, Braak P. Prevalence and characteristics of dementia in Parkinson
E. Staging of brain pathology related to sporadic disease: an 8-year prospective study. Arch Neurol
Parkinson’s disease. Neurobiol Aging 2003;24(2):197–211. 2003;60(3):387–92.
[39] Maetzler W, Liepelt I, Berg D. Progression of Parkinson’s [57] de Lau LM, Schipper CM, Hofman A, Koudstaal PJ, Breteler
disease in the clinical phase: potential markers. Lancet MM. Prognosis of Parkinson disease: risk of dementia and
Neurol 2009;8(12):1158–71. mortality: the Rotterdam Study. Arch Neurol
[40] Auyeung M, Tsoi TH, Mok V, Cheung CM, Lee CN, Li R, 2005;62(8):1265–9.
et al. Ten-year survival and outcomes in a prospective [58] Hobson P, Meara J. Risk and incidence of dementia in a
cohort of new onset Chinese Parkinson’s disease patients. cohort of older subjects with Parkinson’s disease in the
J Neurol Neurosurg Psychiatry 2012;83(6):607–11. United Kingdom. Mov Disord 2004;19(9):1043–9.
[41] Diem-Zangerl A, Seppi K, Wenning GK, Trinka E, [59] Perez F, Helmer C, Foubert-Samier A, Auriacombe S,
Ransmayr G, Oberaigner W, et al. Mortality in Parkinson’s Dartigues JF, Tison F. Risk of dementia in an elderly
disease: a 20-year follow-up study. Mov Disord population of Parkinson’s disease patients: a 15-year
2009;24(6):819–25. population-based study. Alzheimers Dement
[42] Marras C, McDermott MP, Rochon PA, Tanner CM, Naglie 2012;8(6):463–9.
G, Rudolph A, et al. Survival in Parkinson disease: 13-year [60] Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG. The
follow-up of the DATATOP cohort. Neurology Sydney multicenter study of Parkinson’s disease: the
2005;64(1):87–93. inevitability of dementia at 20 years. Mov Disord
[43] Williams-Gray CH, Mason SL, Evans JR, Foltynie T, 2008;23(6):837–44.
Brayne C, Robbins TW, et al. The CamPaIGN study of [61] Aarsland D, Bronnick K, Ehrt U, De Deyn PP, Tekin S, Emre
Parkinson’s disease: 10-year outlook in an incident M, et al. Neuropsychiatric symptoms in patients with
population-based cohort. J Neurol Neurosurg Psychiatry Parkinson’s disease and dementia: frequency, profile and
2013;84(11):1258–64. associated care giver stress. J Neurol Neurosurg Psychiatry
[44] Hobson P, Meara J, Ishihara-Paul L. The estimated life 2007;78(1):36–42.
expectancy in a community cohort of Parkinson’s disease [62] Buter TC, van den Hout A, Matthews FE, Larsen JP, Brayne
patients with and without dementia, compared with the C, Aarsland D. Dementia and survival in Parkinson
UK population. J Neurol Neurosurg Psychiatry disease: a 12-year population study. Neurology
2010;81(10):1093–8. 2008;70(13):1017–22.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 11

[63] Burn DJ, Rowan EN, Allan LM, Molloy S, O’Brien JT, [83] Alves G, Kurz M, Lie SA, Larsen JP. Cigarette smoking in
McKeith IG. Motor subtype and cognitive decline in Parkinson’s disease: influence on disease progression.
Parkinson’s disease. Parkinson’s disease with dementia, Mov Disord 2004;19(9):1087–92.
and dementia with Lewy bodies. J Neurol Neurosurg [84] Morens DM, Grandinetti A, Reed D, White LR, Ross GW.
Psychiatry 2006;77(5):585–9. Cigarette smoking and protection from Parkinson’s
[64] Williams-Gray CH, Foltynie T, Brayne CE, Robbins TW, disease: false association or etiologic clue? Neurology
Barker RA. Evolution of cognitive dysfunction in an 1995;45:1041–51.
incident Parkinson’s disease cohort. Brain [85] Ritz B, Rhodes SL. After half a century of research on
2007;130(7):1787–98. smoking and PD, where do we go now? Neurology
[65] Bajaj A, Driver JA, Schernhammer ES. Parkinson’s disease 2010;74(11):870–1.
and cancer risk: a systematic review and meta-analysis. [86] Ritz B, Lee PC, Lassen CF, Arah OA. Parkinson disease and
Cancer Causes Control 2010;21(5):697–707. smoking revisited: ease of quitting is an early sign of the
[66] Wirdefeldt K, Weibull CE, Chen H, Kamel F, Lundholm C, disease. Neurology 2014;83(16):1396–402.
Fang F, et al. Parkinson’s disease and cancer: a register- [87] Poewe W, Gerstenbrand F, Ransmayr G, Pldrer S.
based family study. Am J Epidemiol 2014;179(1):85–94. Premorbid personality of Parkinson patients. J Neural
[67] Xu L, Lin DC, Yin D, Koeffler HP. An emerging role of Transm 1983;19(Suppl.):215–24.
PARK2 in cancer. J Mol Med (Berl) 2014;92(1):31–42. [88] Menza MA, Golbe LI, Cody RA, Forman NE. Dopamine-
[68] Liu R, Gao X, Lu Y, Chen H. Meta-analysis of the related personality traits in Parkinson’s disease.
relationship between Parkinson disease and melanoma. Neurology 1993;43(3 Pt 1):505–8.
Neurology 2011;76(23):2002–9. [89] Arabia G, Grossardt BR, Colligan RC, Bower JH, Maraganore
[69] Dong J, Gao J, Nalls M, Gao X, Huang X, Han J, et al. DM, Ahlskog JE, et al. Novelty seeking and introversion do
Susceptibility loci for pigmentation and melanoma in not predict the long-term risk of Parkinson disease.
relation to Parkinson’s disease. Neurobiol Aging Neurology 2010;75(4):349–57.
2014;35(6):1512–610. [90] Ishihara L, Brayne C. What is the evidence for a premorbid
[70] Thacker EL, Ascherio A. Familial aggregation of parkinsonian personality: a systematic review. Mov
Parkinson’s disease: a meta-analysis. Mov Disord Disord 2006;21(8):1066–72.
2008;23(8):1174–83. [91] Bower JH, Grossardt BR, Maraganore DM, Ahlskog JE,
[71] Verstraeten A, Theuns J, Van BC. Progress in unraveling Colligan RC, Geda YE, et al. Anxious personality predicts
the genetic etiology of Parkinson disease in a genomic era. an increased risk of Parkinson’s disease. Mov Disord
Trends Genet 2015;31(3):140–9. 2010;25(13):2105–13.
[72] Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad [92] Wirdefeldt K, Gatz M, Pawitan Y, Pedersen NL. Risk and
M, et al. Large-scale meta-analysis of genome-wide protective factors for Parkinson’s disease: a study in
association data identifies six new risk loci for Parkinson’s Swedish twins. Ann Neurol 2005;57(1):27–33.
disease. Nat Genet 2014;46(9):989–93. [93] Tanner CM, Goldman SM, Aston DA, Ottman R, Ellenberg J,
[73] Escott-Price V, Nalls MA, Morris HR, Lubbe S, Brice A, Mayeux R, et al. Smoking and Parkinson’s disease in
Gasser T, et al. Polygenic risk of Parkinson disease is twins. Neurology 2002;58(4):581–8.
correlated with disease age at onset. Ann Neurol [94] Mellick GD, Gartner CE, Silburn PA, Battistutta D.
2015;77(4):582–91. Passive smoking and Parkinson disease. Neurology
[74] Nalls MA, Escott-Price V, Williams NM, Lubbe S, Keller MF, 2006;67(1):179–80.
Morris HR, et al. Genetic risk and age in Parkinson’s [95] Searles Nielsen S, Gallagher LG, Lundin JI, Longstreth Jr
disease: Continuum not stratum. Mov Disord WT, Smith-Weller T, Franklin GM, et al. Environmental
2015;30(6):850–4. tobacco smoke and Parkinson’s disease. Mov Disord
[75] Tanner CM, Ottman R, Goldman SM, Ellenberg J, Chan P, 2012;27(2):293–6.
Mayeux R, et al. Parkinson disease in twins – An etiologic [96] Searles Nielsen S, Franklin GM, Longstreth WT, Swanson
study. JAMA 1999;281(4):341–6. PD, Checkoway H. Nicotine from edible Solanaceae and
[76] Wirdefeldt K, Gatz M, Reynolds CA, Prescott CA, Pedersen risk of Parkinson disease. Ann Neurol 2013;74(3):472–7.
NL. Heritability of Parkinson disease in Swedish twins: a [97] Fowler JS, Volkow ND, Wang GJ, Pappas N, Logan J,
longitudinal study. Neurobiol Aging 2011;32(10). 1923.e1–8. MacGregor R, et al. Inhibition of monoamine oxidase B in
[77] Savica R, Rocca WA, Ahlskog JE. When does Parkinson the brains of smokers. Nature 1996;379(6567):733–6.
disease start? Arch Neurol 2010;67(7):798–801. [98] Ross GW, Petrovitch H. Current evidence for
[78] Wirdefeldt K, Adami HO, Cole P, Trichopoulos D, Mandel J. neuroprotective effects of nicotine and caffeine against
Epidemiology and etiology of Parkinson’s disease: a review Parkinson’s disease. Drug Aging 2001;18(11):797–806.
of the evidence. Eur J Epidemiol 2011;26(Suppl. 1):1–58. [99] Hong DP, Fink AL, Uversky VN. Smoking and Parkinson’s
[79] Hernan MA, Takkouche B, Caamano-Isorna F, Gestal- disease: does nicotine affect alpha-synuclein fibrillation?
Otero JJ. A meta-analysis of coffee drinking, cigarette Biochim Biophys Acta 2009;1794(2):282–90.
smoking, and the risk of Parkinson’s disease. Ann Neurol [100] Schwarzschild MA, Xu K, Oztas E, Petzer JP, Castagnoli
2002;52(3):276–84. K, Castagnoli Jr N, et al. Neuroprotection by caffeine and
[80] Ritz B, Ascherio A, Checkoway H, Marder KS, Nelson LM, more specific A2A receptor antagonists in animal
Rocca WA, et al. Pooled analysis of tobacco use and risk of models of Parkinson’s disease. Neurology 2003;
Parkinson disease. Arch Neurol 2007;64(7):990–7. 61(11 Suppl. 6):S55–61.
[81] Chen H, Huang X, Guo X, Mailman RB, Park Y, Kamel F, [101] Saaksjarvi K, Knekt P, Rissanen H, Laaksonen MA,
et al. Smoking duration, intensity, and risk of Parkinson Reunanen A, Mannisto S. Prospective study of coffee
disease. Neurology 2010;74(11):878–84. consumption and risk of Parkinson’s disease. Eur J Clin
[82] Galanaud JP, Elbaz A, Clavel J, Vidal JS, Corrèze JR, Nutr 2008;62(7):908–15.
Alpérovitch A, et al. Cigarette smoking and Parkinson’s [102] Costa J, Lunet N, Santos C, Santos J, Vaz-Carneiro A.
disease: a case-control study in a population characterized Caffeine exposure and the risk of Parkinson’s disease: a
by a high prevalence of pesticide exposure. Mov Disord systematic review and meta-analysis of observational
2005;20(2):181–9. studies. J Alzheimers Dis 2010;20(Suppl. 1):S221–38.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

12 revue neurologique xxx (2015) xxx–xxx

[103] Ross GW, Abbott RD, Petrovitch H, Morens DM, [123] Fang F, Chen H, Feldman AL, Kamel F, Ye W, Wirdefeldt K.
Grandinetti A, Tung KH, et al. Association of coffee and Head injury and Parkinson’s disease: a population-based
caffeine intake with the risk of Parkinson disease. JAMA study. Mov Disord 2012;27(13):1632–5.
2000;283(20):2674–9. [124] Goldman SM, Kamel F, Ross GW, Jewell SA, Bhudhikanok
[104] Ascherio A, Chen H, Schwarzschild MA, Zhang SM, Colditz GS, Umbach D, et al. Head injury, alpha-synuclein Rep1,
GA, Speizer FE. Caffeine, postmenopausal estrogen, and and Parkinson’s disease. Ann Neurol 2012;71(1):40–8.
risk of Parkinson’s disease. Neurology 2003;60(5):790–5. [125] Lock EA, Zhang J, Checkoway H. Solvents and Parkinson
[105] Antonini A, Poewe W. Adenosine A2A receptor disease: a systematic review of toxicological and
antagonists in Parkinson’s disease: still in the running. epidemiological evidence. Toxicol Appl Pharmacol
Lancet Neurol 2014;13(8):748–9. 2013;266(3):345–55.
[106] Pinna A. Adenosine A2A receptor antagonists in [126] Feldman AL, Johansson AL, Nise G, Gatz M, Pedersen NL,
Parkinson’s disease: progress in clinical trials from the Wirdefeldt K. Occupational exposure in Parkinsonian
newly approved istradefylline to drugs in early disorders: a 43-year prospective cohort study in men.
development and those already discontinued. CNS Drugs Parkinsonism Relat Disord 2011;17(9):677–82.
2014;28(5):455–74. [127] Goldman SM, Quinlan PJ, Ross GW, Marras C, Meng C,
[107] van der Mark M, Brouwer M, Kromhout H, Nijssen P, Huss Bhudhikanok GS, et al. Solvent exposures and Parkinson
A, Vermeulen R. Is pesticide use related to Parkinson’s disease risk in twins. Ann Neurol 2012;71(6):776–84.
disease? Some clues to heterogeity in study results. [128] Gash DM, Rutland K, Hudson NL, Sullivan PG, Bing G, Cass
Environ Health Perspect 2012;120(3):340–7. WA, et al. Trichloroethylene: Parkinsonism and complex 1
[108] Van Maele-Fabry G, Hoet P, Vilain F, Lison D. Occupational mitochondrial neurotoxicity. Ann Neurol 2008;63(2):
exposure to pesticides and Parkinson’s disease: a 184–192.
systematic review and meta-analysis of cohort studies. [129] Morelli M, Carta AR, Kachroo A, Schwarzschild MA.
Environ Int 2012;46:30–43. Pathophysiological roles for purines: adenosine, caffeine
[109] Pezzoli G, Cereda E. Exposure to pesticides or solvents and and urate. Prog Brain Res 2010;183:183–208.
risk of Parkinson disease. Neurology 2013;80(22):2035–41. [130] Shen C, Guo Y, Luo W, Lin C, Ding M. Serum urate and the
[110] Moisan F, Spinosi J, Delabre L, Gourlet V, Mazurie JL, risk of Parkinson’s disease: results from a meta-analysis.
Benatru I, et al. Association of Parkinson’s disease and its Can J Neurol Sci 2013;40(1):73–9.
subtypes with agricultural pesticide exposures in men: a [131] Gonzalez-Aramburu I, Sanchez-Juan P, Sierra M,
case-control study in France. Environ Health Perspect Fernandez-Juan E, Sanchez-Quintana C, Berciano J, et al.
2015. http://dx.doi.org/10.1289/ehp.1307970. Serum uric acid and risk of dementia in Parkinson’s
[111] Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, disease. Parkinsonism Relat Disord 2014;20(6):637–9.
Korell M, et al. Rotenone, Paraquat and Parkinson’s [132] Simon KC, Eberly S, Gao X, Oakes D, Tanner CM, Shoulson
disease. Environ Health Perspect 2011;119(6):866–72. I, et al. Mendelian randomization of serum urate and
[112] Elbaz A, Clavel J, Rathouz PJ, Moisan F, Galanaud JP, parkinson disease progression. Ann Neurol 2014;76(6):
Delemotte B, et al. Professional exposure to pesticides and 862–868.
Parkinson’s disease. Ann Neurol 2009;66(4):494–504. [133] Schwarzschild MA, Ascherio A, Beal MF, Cudkowicz ME,
[113] Costello S, Cockburn M, Bronstein J, Zhang X, Ritz B. Curhan GC, Hare JM, et al. Inosine to increase serum and
Parkinson’s disease and residential exposure to maneb cerebrospinal fluid urate in Parkinson disease: a
and paraquat from agricultural applications in the central randomized clinical trial. JAMA Neurol 2014;71(2):141–50.
valley of California. Am J Epidemiol 2009;169(8):919–26. [134] Chen H, O’Reilly E, McCullough ML, Rodriguez C,
[114] Wang A, Cockburn M, Ly TT, Bronstein JM, Ritz B. The Schwarzschild MA, Calle EE, et al. Consumption of dairy
association between ambient exposure to products and risk of Parkinson’s disease. Am J Epidemiol
organophosphates and Parkinson’s disease risk. Occup 2007;165(9):998–1006.
Environ Med 2014;71(4):275–81. [135] Kyrozis A, Ghika A, Stathopoulos P, Vassilopoulos D,
[115] Wang A, Costello S, Cockburn M, Zhang X, Bronstein J, Ritz Trichopoulos D, Trichopoulou A. Dietary and lifestyle
B. Parkinson’s disease risk from ambient exposure to variables in relation to incidence of Parkinson’s disease in
pesticides. Eur J Epidemiol 2011;26(7):547–55. Greece. Eur J Epidemiol 2013;28(1):67–77.
[116] Hatcher JM, Pennell KD, Miller GW. Parkinson’s disease [136] Tansey MG, Frank-Cannon TC, McCoy MK, Lee JK,
and pesticides: a toxicological perspective. Trends Martinez TN, McAlpine FE, et al. Neuroinflammation in
Pharmacol Sci 2008;29(6):322–9. Parkinson’s disease: is there sufficient evidence for
[117] Expertise collective. Pesticides : effets sur la santé. Inserm mechanism-based interventional therapy? Front Biosci
2013. 2008;13:709–17.
[118] Baltazar MT, Dinis-Oliveira RJ, de Lourdes BM, Tsatsakis [137] Hartmann A, Hunot S, Hirsch EC. Inflammation and
AM, Duarte JA, Carvalho F. Pesticides exposure as dopaminergic neuronal loss in Parkinson’s disease: a
etiological factors of Parkinson’s disease and other complex matter. Exp Neurol 2003;184(2):561–4.
neurodegenerative diseases – A mechanistic approach. [138] Kieburtz K, Wunderle KB. Parkinson’s disease: evidence
Toxicol Lett 2014;230(2):85–103. for environmental risk factors. Mov Disord 2013;28(1):8–13.
[119] Jafari S, Etminan M, Aminzadeh F, Samii A. Head injury [139] Gagne JJ, Power MC. Anti-inflammatory drugs and risk of
and risk of Parkinson disease: a systematic review and Parkinson disease: a meta-analysis. Neurology
meta-analysis. Mov Disord 2013;28(9):1222–9. 2010;74(12):995–1002.
[120] Goldman SM, Tanner CM, Oakes D, Bhudhikanok GS, [140] Chaturvedi RK, Beal MF. PPAR: a therapeutic target in
Gupta A, Langston JW. Head injury and Parkinson’s Parkinson’s disease. J Neurochem 2008;106(2):506–18.
disease risk in twins. Ann Neurol 2006;60:65–72. [141] de Lau LM, Koudstaal PJ, Hofman A, Breteler MM. Serum
[121] Harris MA, Shen H, Marion SA, Tsui JK, Teschke K. Head cholesterol levels and the risk of Parkinson’s disease. Am J
injuries and Parkinson’s disease in a case-control study. Epidemiol 2006;164(10):998–1002.
Occup Environ Med 2013;70(12):839–44. [142] Huang X, Alonso A, Guo X, Umbach DM, Lichtenstein ML,
[122] Bower JH, Maraganore DM, Peterson BJ, McDonnell SK, Ballantyne CM, et al. Statins, plasma cholesterol, and risk
Ahlskog JE, Rocca WA. Head trauma preceding PD – A of Parkinson’s disease: a prospective study. Mov Disord
case-control study. Neurology 2003;60(10):1610–5. 2015;30(4):552–9.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012
NEUROL-1580; No. of Pages 13

revue neurologique xxx (2015) xxx–xxx 13

[143] Simon KC, Chen H, Schwarzschild M, Ascherio A. [152] Lee PC, Rhodes SL, Sinsheimer JS, Bronstein J, Ritz B.
Hypertension, hypercholesterolemia, diabetes, and risk of Functional paraoxonase 1 variants modify the risk of
Parkinson disease. Neurology 2007;69(17):1688–95. Parkinson’s disease due to organophosphate exposure.
[144] Huang X, Auinger P, Eberly S, Oakes D, Schwarzschild M, Environ Int 2013;56:42–7.
Ascherio A, et al. Serum cholesterol and the progression of [153] Elbaz A, Levecque C, Clavel J, Vidal JS, Richard F, Amouyel
Parkinson’s disease: results from DATATOP. PLoS One P, et al. CYP2D6 polymorphism, pesticide exposure, and
2011;6(8):e22854. Parkinson’s disease. Ann Neurol 2004;55(3):430–4.
[145] Undela K, Gudala K, Malla S, Bansal D. Statin use and risk [154] Fitzmaurice AG, Rhodes SL, Cockburn M, Ritz B, Bronstein
of Parkinson’s disease: a meta-analysis of observational JM. Aldehyde dehydrogenase variation enhances effect of
studies. J Neurol 2013;260(1):158–65. pesticides associated with Parkinson disease. Neurology
[146] Lee YC, Lin CH, Wu RM, Lin MS, Lin JW, Chang CH, et al. 2014;82(5):419–26.
Discontinuation of statin therapy associates with [155] Rhodes SL, Fitzmaurice AG, Cockburn M, Bronstein JM,
Parkinson disease: a population-based study. Neurology Sinsheimer JS, Ritz B. Pesticides that inhibit the ubiquitin-
2013;81(5):410–6. proteasome system: effect measure modification by
[147] Chan CS, Guzman JN, Ilijic E, Mercer JN, Rick C, Tkatch T, genetic variation in SKP1 in Parkinsons disease. Environ
et al. ‘‘Rejuvenation’’ protects neurons in mouse models Res 2013;126:1–8.
of Parkinson’s disease. Nature 2007;447(7148):1081–6. [156] Hamza TH, Chen H, Hill-Burns EM, Rhodes SL,
[148] Lang Y, Gong D, Fan Y. Calcium channel blocker use and Montimurro J, Kay DM, et al. Genome-wide gene-
risk of Parkinson’s disease: a meta-analysis. environment study identifies glutamate receptor gene
Pharmacoepidemiol Drug Saf 2015;24(6):559–66. GRIN2A as a Parkinson’s disease modifier gene via
[149] Lee YC, Lin CH, Wu RM, Lin JW, Chang CH, Lai MS. interaction with coffee. PLoS Genet 2011;7(8):e1002237.
Antihypertensive agents and risk of Parkinson’s disease: a [157] Ahmed I, Lee PC, Lill CM, Searles NS, Artaud F, Gallagher
nationwide cohort study. PLoS One 2014;9(6):e98961. LG, et al. Lack of replication of the GRIN2A-by-Coffee
[150] Parkinson Study Group. Phase II safety, tolerability, and interaction in Parkinson disease. PLoS Genet
dose selection study of isradipine as a potential disease- 2014;10(11):e1004788.
modifying intervention in early Parkinson’s disease [158] Feng Y, Jankovic J, Wu YC. Epigenetic mechanisms in
(STEADY-PD). Mov Disord 2013;28(13):1823–31. Parkinson’s disease. J Neurol Sci 2015;349(1–2):3–9.
[151] Dutheil F, Beaune P, Tzourio C, Loriot MA, Elbaz A. [159] Searles NS, Checkoway H, Butler RA, Nelson HH, Farin
Interaction between ABCB1 and professional exposure to FM, Longstreth Jr WT, et al. LINE-1 DNA methylation,
organochlorine insecticides in Parkinson disease. Arch smoking and risk of Parkinson’s disease. J Parkinsons Dis
Neurol 2010;67(6):739–45. 2012;2(4):303–8.

Please cite this article in press as: Elbaz A, et al. Epidemiology of Parkinson’s disease. Revue neurologique (2015), http://dx.doi.org/10.1016/
j.neurol.2015.09.012

You might also like