Semin Neurol 2017; 37(02): 135-146
DOI: 10.1055/s-0037-1601567
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Genetic Forms of Parkinson's Disease

Christine Y. Kim
2   Department of Movement Disorders, Columbia University Medical Center, New York, New York
,
Roy N. Alcalay
1   Department of Neurology, Columbia University Medical Center, New York, New York
› Author Affiliations
Further Information

Address for correspondence

Roy N. Alcalay, MD, MS
Department of Neurology, Columbia University Medical Center
710 W 168th St, 3rd floor, New York
NY 10032

Publication History

Publication Date:
16 May 2017 (online)

 

Abstract

One of the greatest advances in Parkinson's disease (PD) research in the past two decades has been a better understanding of PD genetics. Of the many candidate genes investigated, the best studied include LRRK2, SNCA, VPS35, Parkin, PINK1, and DJ1. The authors review the key clinical features of these monogenic forms, as well as for the prevalent risk factor gene, GBA, including the phenotype, clinical course, and treatment response. They also outline areas for future investigation: longitudinal studies of PD's clinical course, the identification of its premotor manifestations, and its specific mechanisms of pathogenicity.


#

The clinical course and treatment response of Parkinson's disease (PD) are extremely heterogeneous, creating challenges in its management. Similarly, PD's etiology is heterogeneous, multifactorial, and often complex. Nevertheless, our understanding of the disease's genetic causes and risk factors has undergone substantial development in the last 20 years. The genetic risks for PD can be divided into those associated with a high risk for PD and those that increase the PD risk only modestly. Mutations associated with a high risk for PD are often called monogenic, Mendelian, or causative (from hereon, causative) and underlie 5 to 10% of PD cases.[1] Causative mutations are rarer than risk factors. In some cases, different alterations in the same gene are causative, whereas others are considered risk factors. A classic example is the gene SNCA, which encodes α-synuclein. A rare A53T mutation in the gene causes PD in the majority of carriers, but a single nucleotide polymorphism in the vicinity of the gene is a known risk factor replicated in multiple genome-wide association studies. Here we will review the clinical features of the major causative genes: LRRK2, SNCA, VPS35, Parkin, PINK1, and DJ1, as well as GBA.

LRRK2

History

An autosomal dominant PD (ADPD) cohort was first described among a Nebraska kindred in 1995.[2] The locus 12p11.21 was then identified via a genome-wide linkage analysis of a Japanese family with ADPD in 2002.[3] Through recombination mapping and candidate gene sequencing, the LRRK2 gene was identified in 2004.[4] [5] Subsequently, multiple pathogenic mutations have been identified, with ethnic distribution and pathogenicity varying by the specific mutation.


#

Mechanism of Pathogenicity

Our current understanding of LRRK2 function, interactions, and pathogenicity in PD is incomplete. LRRK2 contains multiple functional domains, including a kinase domain. It is believed that increased LRRK2 activity increases the risk for PD because increased kinase activity has been associated with nigrostriatal degeneration and Lewy body (LB) formation. In addition, the G2019S mutation, located in the kinase domain, has been associated with increased phosphorylation activity in vivo.[6] However, the pathogenicity of other mutations may be mediated by other mechanisms. Furthermore, the substrates of LRRK2 phosphorylation (except for autophosphorylation) were unknown until recently. Just recently identified as substrates were the Rab GTPases (Rab5 and Rab7), which affect signaling cascades, degradation, and endosomal trafficking.[6] [7] Abnormal mitochondrial morphology and function have been noted among LRRK2 carriers, as well as an abnormal accumulation in autophagic vacuoles, thought to be linked via the regulatory protein 5′ AMP-activated protein kinase.[6]


#

Frequency and Ethnic Distribution

Ethnic distribution varies widely by mutation. The most common mutation,[8] G2019S, is especially common in North African Berbers and Ashkenazi Jews (AJs). Mutations in the 1441 nucleotide are more common in Spain among the Basque population.[9] Other mutations are implicated in Asian populations. The I2020T mutation has been reported in Japanese cohorts,[10] although the mutation ultimately appears to be rare among other Asian populations.[11] A common variant in Asian populations, G2385R, has been identified as a risk factor in Chinese, Japanese, and Korean populations.[12] [13] [14] Among Han Chinese populations, it has been reported with a frequency of 8 to 11.7% among PD cohorts and 0.5 to 3.3% in the general population.[15] [16] [17] The mutation frequency appears similar in a Korean population.[13] Few studies have compared the PD phenotype with that of other mutations, but the motor phenotype may be more severe than for G2019S PD; G2385R PD patients have been noted to have higher Unified Parkinson's Disease Rating Scale (UPDRS) motor scores with more frequent fluctuations than G2019S PD or idiopathic PD (iPD) patients.[18]


#

G2019S

Of all the LRRK2 mutations, the best described is G2019S. The mutation has been identified among PD patients in multiple ethnic groups including Norwegians,[19] Italians, Portuguese, and Brazilians.[20] In a large worldwide study, mutation frequency was found to be 1% among sporadic and 4% among familial PD patients, but with the highest mutation frequency among North African Arab and Ashkenazi Jewish populations: 39% and 38% among North African Arabs, and 10% and 28% among Ashkenazi Jews with sporadic and familial PD, respectively.[21] It is apparently rare in Asian populations.[22] Penetrance estimations vary widely and range between 25 to 100% by 80 years.[21] [23]


#

Phenotype

Overall, the G2019S motor phenotype appears to overlap with that of iPD.[19] However, carriers more commonly manifest a postural instability gait difficulty phenotype than do noncarriers (92.3% vs. 58.9%).[24] [25] A good response to levodopa has been reported,[26] although it has been associated with levodopa-induced dyskinesias (LIDs).[27] Regarding the rate of progression, a longitudinal study by Nabli et al found similar rates of progression in the UPDRS and Hoehn and Yahr Scale scores between G2019S and iPD cohorts at 6-year follow-up.[25]

G2019S PD patients have fewer nonmotor manifestations than do noncarriers. Ben Sassi et al found similar cognitive involvement in carrier versus noncarrier PD patients.[28] In a larger study, Alcalay et al reported superior performance in attention, executive function, and language domains in G2019S PD patients versus iPD patients, despite longer disease duration in the G2019S cohort.[29] G2019S PD patients appear to have less depression[18] and less hyposmia than iPD patients,[30] [31] [32] and less probable rapid eye movement sleep behavior disorder (RBD), as assessed by questionnaire, than iPD patients.[30] [33]

LRRK2's potential gain of function mechanism has raised concern for a possible increased risk of malignancy. An increased risk of nonskin cancers among G2019S carriers versus noncarriers has been reported in AJ PD patients[34] [35] and requires further longitudinal study.


#

Premotor State

Much effort is invested in identifying carriers without motor PD who may develop PD. A subset of nonmanifesting carriers may have impaired olfaction compared with noncarrier controls, suggesting a possible preclinical marker for PD, although longitudinal studies are lacking.[32] [36] In a cross-sectional study, Mirelman et al found that the most prominent clinical difference between carriers of G2019S and noncarriers was reduced arm swing when walking.[37] [38] One study found an increased risk of premorbid mood disorders (odds ratio, 6) in carriers versus noncarriers, although there were no differences found in neuropsychological testing.[39]


#

Pathology

Postmortem examination is notable for the degeneration of the substantia nigra (SN) and locus coeruleus (LC), but with variable LB and tau pathology. The initial report by Zimprich et al of Y1699C and R1441C mutation carriers noted diverse pathology including LB PD, diffuse LB disease, and nigral degeneration, as well as progressive supranuclear palsy-like pathology.[4] Subsequent examinations reported variable cortical LB pathology, redemonstrated through two subsequent case series. Upon review of the postmortem examination of 28 G2019S PD patients, Poulopoulos et al reported SN and LC neuronal loss in all cases that had parkinsonism, with LB pathology in a majority but with variable cortical involvement. They also noted tau pathology in a majority, but with variable location and severity. Interestingly, PD patients with mutations other than G2019S had a lower frequency of LB pathology and more SN versus LC degeneration, suggesting mutation-specific pathogenicity.[40] Upon review of the postmortem examination of 37 LRRK2 PD patients with corresponding clinical data, Kalia et al found neuronal SN loss in all cases, redemonstrating LB variability. Of note, on clinicopathological correlation, a primarily motor phenotype was associated with an absence of LBs, and nonmotor features were associated with the presence of LBs.[41]


#
#

SNCA

History

Alpha synuclein (SNCA) mutations have emerged as a rare, but important cause of ADPD with high penetrance. Polymeropoulos et al first identified an Italian kindred with ADPD with an iPD phenotype, but with heterogeneous age-at-onset, which was associated with a locus at 4q21-23 in 1996 through linkage analysis.[42] Subsequently, a mutation in SNCA, A53T, was identified,[43] but with apparently rare frequency based on early mutation screening among iPD cohorts of early or late-onset PD.[44] [45] Four other missense mutations have since been identified. A30P and E46K missense mutations were identified in German and Spanish kindreds, respectively.[46] [47] More recently, a H50Q point mutation has been reported in a British family.[48] A G51D mutation was reported in a French family with atypical PD, with prominent psychiatric symptoms and associated pyramidal signs. The postmortem examination was notable for cytoplasmic inclusions in the pyramidal tracts as well as in the basal ganglia.[49] Overall, in comparison with iPD, the SNCA PD clinical course is notable for earlier age of onset and more rapid progression, with good levodopa response, but early motor fluctuations.[50] Severe depression, including completed suicide, has been reported.[51]

Of the missense mutations, A53T is most common, although still rare overall, with only ∼70 reported cases, with an apparently aggressive clinical course, notable for a 10-year earlier age-at-onset than the other missense mutations.[50] Recently, Papadimitriou et al performed a prospective 2-year longitudinal follow-up among A53T symptomatic and asymptomatic carriers, noting prominent motor and nonmotor decline that included olfactory, autonomic, and cognitive dysfunction, with a disease penetrance of ∼90%.[52]


#

Mechanism of Pathogenicity

The mechanism of pathogenicity remains unclear, but given that SNCA-associated PD demonstrates a gene dosage effect, a gain of function is suspected. Gene multiplication appears to be more common among European and Asian populations. Triplication has been associated with an earlier onset of disease, a more severe phenotype with some atypical features including myoclonus, and more rapid progression than in duplication.[50] A case of gene triplication reported by Singleton et al had a clinical course notable for early onset, and a postmortem examination notable for prominent cortical and subcortical LB pathology.[53] Duplications have also been reported in multiple ADPD families,[54] as well as in sporadic PD.[55] [56] Phenotypes reported include prominent psychiatric symptoms including visual hallucinations.[54]


#
#

VPS35

The VPS35 gene was identified in 2011 among a Swiss kindred with late-onset ADPD via next-generation sequencing[57] and an Austrian ADPD kindred via exome sequencing.[58] In population studies, the mutation frequency appears rare, with one study of 475 patients with familial PD finding no cases of D620N mutation.[59] Other studies among European and Asian populations have found a frequency of ∼1% in ADPD, with Lesage et al finding the D620N mutation in 3 of 246 ADPD patients and not in controls.[60] A similar frequency was reported in a Japanese ADPD population; the motor phenotype in that study was tremor-predominant PD.[61] However, larger phenotype-focused studies are lacking.


#

Parkin

History

Parkin mutations are the most common cause of autosomal recessive PD and are especially prevalent in PD with onset before age 30. Ishikawa et al characterized a cohort of patients with familial juvenile parkinsonism in 1996, noting female predominance with young onset (mean age-at-onset 27.8 years) and slow progression. The phenotype was notable for relatively mild tremor, rigidity, and bradykinesia, with some atypical features including prominent freezing of gait, retropulsion, and foot dystonia, as well as hyperreflexia, with notable sleep benefit on most symptoms; no dementia or autonomic features were noted. An excellent response with levodopa was noted, but with frequent LID and wearing off.[62] Matsumine et al identified an associated locus at 6q25.2–27 through linkage analysis.[63] Kitada et al then identified the Parkin gene and protein in 1998.[64] Hattori et al identified four homozygous deletional mutations.[65] It was subsequently implicated in sporadic cases[66] and is now confirmed in multiple ethnic groups including European, Hispanic, African American, and North African.[67] [68] [69]


#

Mechanism of Pathogenicity

The mechanism of pathogenicity remains unclear. Parkin is an E3 ubiquitin ligase protein, catalyzing the transfer of ubiquitin to its specific target protein. Multiple target proteins with widely variable functions have been suggested as possible Parkin substrates. A role in protein targeting for proteasomal degradation has been proposed.[70] In one capacity, Parkin appears to work with PINK1 (see below) in organellar quality control through the activation of mitophagy in the setting of mitochondrial damage.[71] Finding the mechanism of the specific pathological changes of PD requires continued investigation, but given that most Parkin postmortem studies (detailed below) do not demonstrate α-synuclein pathology, the mechanism may be different than iPD.


#

Frequency

Parkin mutations are the most common cause of early-onset PD, defined in different studies as onset before age 40 to 51 years. The reported mutation frequency has varied widely. In a meta-analysis of studies among early-onset PD subjects, Kilarski et al found a mutation frequency of 15.5% among familial (although significantly higher in known consanguineous cases at 31.4%) and 4.3% among sporadic cases.[72] However, at least 60 mutations and variants have been identified, raising significant challenges in determining pathogenicity, which may vary with specific mutation.[73] In addition, deletions and duplications are especially common, which complicate Parkin genotyping. Such alterations may not be identified in genome-wide association studies or whole-exome sequencing, and in addition to Sanger sequencing, a dosage analysis is required to determine if deletions or duplications are present. Allele frequency may be ethnically dependent.[74] Marder et al found an increased risk of Parkin mutation (odds ratio, 2.8) among Hispanic versus non-Hispanic white early-onset PD patients.[69]


#

Phenotype

The clinical phenotype was first described by Ishikawa et al as above, although phenotypic variability has been noted,[75] in part related to specific mutations. Parkin-associated PD is predominantly early-onset; late-onset cases are established, but are rarer. Cases with later onset (> 45 years old) have been noted in studies including gene dosage analyses,[76] but with lower frequency than in early-onset PD.[77] [78] Disease progression appears to be slower than iPD, also supported by 18F-DOPA positron emission tomography data.[79]


#

Nonmotor Symptoms

Overall, nonmotor symptoms appear less severe in Parkin PD than in iPD. Data regarding cognitive function are cross-sectional and overall suggest comparable, if not superior cognitive function in Parkin PD compared with iPD patients. Several studies have found comparable cognitive function between Parkin PD and iPD groups by neuropsychological testing.[80] [81] A follow-up study by the Consortium on Risk for Early-Onset Parkinson's Disease examined cognitive function in early-onset PD with long-duration disease (>14 years) and found carriers performed better on the Mini-Mental State Examination; were more likely to have lower scores on clinical dementia ratings; and had better attention, memory, and visuospatial performance.[82] Parkin homozygotes and compound heterozygotes with PD may have less olfactory dysfunction than iPD[83] patients or Parkin heterozygotes with PD.[84]

Parkin carriers may, however, have more severe impulse control disorders than noncarriers, which may be a consideration in treatment selection. Morgante et al recently assessed impulse control behaviors in PD biallelic Parkin mutation carriers versus noncarriers, matched for disease duration, age, and dopamine dose equivalent. Although the frequency of at least one impulse control behavior was similar between both groups, Parkin carriers had a higher frequency of compulsive shopping, binge eating, and punding/hobbyism; were more likely to be smokers; and more likely to have higher Questionnaire for Impulsive-Compulsive Disorders in Parkinson's Disease–Rating Scale scores.[85]

Parkin has been proposed to have a tumor-suppressor function in systemic cancers,[86] [87] [88] [89] but this has not been consistently supported in epidemiologic studies. A retrospective study by Alcalay et al did not find a difference in reported cancer history among carriers versus noncarriers.[90]


#

Heterozygotes

It remains uncertain if heterozygous Parkin mutations confer a risk for PD.[77] [91] A few studies suggest that only specific genetic mutations (e.g., duplications, deletions, or point mutations in functionally crucial domains) increase PD risk.[92] [93]


#

Deep Brain Stimulation

Given frequent motor fluctuations and dyskinesias, deep brain stimulation (DBS) may be a relevant treatment consideration for Parkin PD patients. STN DBS appears to have similar efficacy for Parkin PD versus noncarriers. Romito et al performed a post hoc genetic analysis of a series of patients undergoing STN DBS, with a population including Parkin carriers (one compound heterozygote and four heterozygotes) and noncarriers, matched for disease duration and age. The Parkin and non-Parkin groups showed a similar reduction in UPDRS motor scores (56% vs. 51%) with a nonsignificant trend to greater levodopa dose reduction postoperatively in the Parkin group. Behavioral complications were similar, although limited by small numbers.[94] Moro et al performed Parkin and PINK1 mutation screening among 80 patients with early-onset PD who underwent bilateral STN DBS, identifying 11 Parkin mutation carriers (six homozygous or compound heterozygous; five heterozygous) and one PINK1 homozygous mutation carrier. One-year follow-up showed less improvement in mutation carriers by UPDRS motor scores (56% improvement vs. 36% improvement), but this difference was not maintained at the 3- to 6-year follow-up (44% vs. 42%), suggesting mutation carriers benefit from DBS, but not more than iPD patients.[95] Among a cohort of DBS patients screened for PD-associated mutations, Angeli et al found Parkin carriers had the youngest age of onset, but longer disease duration before DBS was required.[96]


#

Pathology

Postmortem examination is notable for SN neuronal loss, but without prominent LB pathology. A review of 77 cases by Pramstaller et al noted neuronal loss in the pigmented nuclei of the brainstem (SN pars compacta [SNpc] and LC), but typical LB pathology in only a portion of cases.[97] More recently, Doherty et al performed a postmortem pathological examination on Parkin PD patients, iPD patients, and controls. Among the Parkin PD cases, they found focal nigral degeneration with ventral predominance and absent or rare LBs, with counts of SNpc demonstrating neuronal loss as severe as in iPD, but with the relative preservation of the dorsal tier, mild neuronal loss in the LC, and the dorsal motor nucleus of vagus but not the nucleus basalis of Meynert, raphe, or other regions. LB pathology was again minimal, with sparse LBs noted in only two cases of five.[98]


#
#

PINK1

History

PINK1 mutations are the second-most common cause of ARPD after Parkin, with some overlap with the Parkin phenotype. The locus was first identified in a Sicilian family (Marsala kindred) with four affected members with early-onset PD, with a course notable for slow progression and a sustained response to levodopa. Through linkage analysis, a novel locus at 1p35-p36 was identified.[99] In 2004, Valente et al identified the associated protein, a mitochondrial kinase, 1 PTEN-induced kinase (PINK1) in three affected families, identifying two homozygous mutations of the kinase domain.[100] Valente et al subsequently noted PINK1 mutations in a cohort of Italian early-onset PD patients.[101] Subsequent studies confirmed PINK1 mutations in North American,[102] other European,[103] and Asian populations.[104] Mutation frequency is reported as ∼4 to 7% in sporadic early-onset PD.[101] [105]


#

Mechanism of Pathogenicity

As first suggested by Valente et al via cell culture studies, PINK1 localizes to the mitochondria,[99] imported there via targeting. PINK1 is a serine/threonine kinase, also containing critical regulatory sites. Mutations conferring a complete loss of kinase activity are associated with early-onset PD.[106] PINK1 functions most prominently together with Parkin in the activation of mitophagy, accumulating on the outer mitochondrial membrane in the setting of mitochondrial damage.[106] The specific mechanism of pathogenicity in PD is currently unclear and requires continued investigation.


#

Phenotype

Phenotype was initially described in the above-mentioned Marsala kindred.[99] Initial studies noted typical parkinsonism of slow progression, with good and persistent levodopa response and minimal cognitive involvement.[101] [107] Later studies noted some atypical features, including dystonia and sleep benefit as seen in Parkin PD,[105] as well as hyperreflexia.[103] Regarding DBS, as discussed above, Moro et al noted a comparable response to iPD patients to STN DBS among their cohort, which included one PINK1 PD patient.[95]


#

Nonmotor Symptoms

Psychiatric features may be present; anxiety and depression have been reported.[108] Hyposmia appears to be a common finding in PINK1 PD. Ferraris et al compared olfaction in iPD, PINK1 homozygous PD, PINK1 heterozygous PD, and PINK1 heterozygous asymptomatic carriers, noting all affected PINK1 subjects and all but one of the PINK1 heterozygotes to be hyposmic.[109]


#

Heterozygotes

Implications for heterozygotes remain controversial, but heterozygosity may be a risk factor for late-onset PD, as suggested by population studies[105] [110] and supported in meta-analysis by Kasten et al.[111] Among affected carriers, the cardinal features and age-at-onset remained similar between the homozygous and heterozygous groups, with a trend toward more gait disturbance in the homozygous group, but in the setting of longer average disease duration.[111] In the preclinical state, heterozygotes may manifest hyposmia.[109]


#

Pathology

Postmortem examinations are limited in number. Postmortem examination has been reported on two PINK1 PD patients with atypical LB pathology. In a compound heterozygous early-onset PD patient, Sammaranch et al noted neuronal loss in the SNpc, with LB pathology in an atypical distribution involving the reticular nuclei, SNpc, and the nucleus of Meynert, but sparing the LC and amygdala.[112] Recently, in a homozygous early-onset PD patient, Takanashi et al found no LB pathology, apart from in a few olfactory nerve neurites, but did note marked SN and LC depigmentation.[113]


#
#

DJ1

van Duijn et al first identified a locus at 1p36, separate from PARK6, in a consanguineous Dutch kindred with early-onset PD.[114] Bonifati et al then identified the DJ1 gene in a Dutch and an Italian family, respectively. The phenotype among the four subjects was early-onset PD (all < 41 years old), one with blepharospasm, with two on treatment with levodopa with good response, and one with motor fluctuation including LID.[115] Mutation frequency is apparently rare, with studies finding a frequency of 0 to 1% in early-onset PD cohorts[116] [117] [118]; a meta-analysis by Kilarski et al found an overall mutation frequency of 0.4%, marginally higher among familial PD (0.8%) than among sporadic PD (0.4%) cases.[72]


#

GBA

History

Clinical observation of parkinsonism among Gaucher's disease (GD) patients led to the confirmation of increased PD prevalence in that population. An initial case series among GD patients with PD described an aggressive phenotype with age-at-onset in the fourth to sixth decade of life, rapid progression, and poor response to levodopa.[119] Aharon-Peretz et al further noted younger onset in carriers than in noncarriers,[120] bringing the GBA gene under investigation as a possible genetic risk factor for PD.


#

Mechanism of Pathogenicity

GBA encodes the lysosomal enzyme glucocerebrosidase (GCase), which cleaves the β-glucosyl linkage of glucosylceramide and glucosylsphingosine. Given the low penetrance of PD, both gain-of-function and loss-of-function mechanisms have been proposed. Proposed gain-of-function mechanisms include facilitation of α − synuclein accumulation by misfolded GCase or lysosomal dysfunction causing impairment in the ubiquitin-proteasome or autophagy pathways. Possible loss-of-function mechanisms include substrate accumulation due to lysosomal dysfunction or altered lipid maintenance.[121] Alcalay et al compared GCase activity in PD patients and controls with and without GBA mutations. Homozygotes and compound heterozygotes had lower enzymatic activity than heterozygotes, who in turn had lower activity than controls. PD patients as a group had lower mean GCase activity than the non-PD group. Further, among noncarriers with PD, lower GCase activity has been associated with shorter disease duration, suggesting lower GCase activity may be associated with faster progression.[122]


#

Frequency

Multiple studies have established a significantly increased risk of PD associated with GBA mutation, now reported worldwide,[120] [123] [124] [125] [126] [127] [128] but most substantially in the AJ population.[129] In the AJ population, mutations in GBA are common among controls. In a large population study, Gan-Or et al found a mutation frequency of 6.35% among young controls (ages 20–45 years) and 4.2% among elderly controls.[129] Penetrance is incomplete and increases with age. It is estimated at 7.7 to 29.7% by 80 years.[130] [131] Further, the conferred risk for PD may increase with functional severity of the GBA mutation. Gan-Or et al found an increased risk for PD conferred with functionally severe versus mild mutations, in keeping with a prior study.[129] [132]


#

Phenotype

Overall, the motor phenotype is similar between GBA PD and iPD,[120] possibly with more bradykinesia and LID among GBA PD patients.[125] In contrast, nonmotor symptoms appear to be more prominent among GBA PD patients than in iPD. One of the largest population studies noted GBA PD patients were more likely than iPD patients to have atypical features including cognitive changes.[128] Greater cognitive involvement has been substantiated in subsequent studies,[133] with Oeda et al also noting possible earlier onset in a retrospective review.[134] Particular impairments in memory and visuospatial domains have been reported.[135] In addition, neuropsychiatric disturbances and autonomic dysfunction may be more severe.[136] GBA mutations have been associated with confirmed idiopathic RBD as well as probable RBD among PD patients.[137] Olfaction appears comparably affected as in iPD.[135]

As was originally reported, the disease course appears to be more rapidly progressive than in iPD. In a longitudinal study of PD patients with and without GBA mutation, Brockmann et al found a more rapid progression of motor as well as cognitive impairment, with decreased survival rates in GBA PD over a 3-year follow-up.[133] Cilia et al also found a decreased survival rate for GBA PD versus iPD patients.[138] Deep brain stimulation may be required earlier in GBA PD, but has also been associated with early cognitive impairment after DBS[96]; further studies are needed.

Biallelic carriers may have more severe disease. Thaler et al recently reported a gene dosage effect, with earlier disease onset and more severe motor and nonmotor symptoms (i.e., cognition, olfaction, RBD, hallucinations) in homozygotes and compound heterozygotes versus both heterozygotes and iPD patients.[139]


#

Pathology

In parallel with GBA PD's prominent cognitive features, cortical LBs have been noted on postmortem examination, although it remains unclear whether they occur with greater burden than in iPD. Clark et al found GBA mutation status to be associated with cortical LBs and not with Alzheimer's disease pathology.[140] In the postmortem examination of 17 GBA PD patients, Neumann et al found widespread, abundant LBs in all, with limbic or diffuse neocortical LB pathology,[126] although with neocortical burden ultimately felt to be comparable to noncarriers.[141] Co-localization of mutant GCase with α − synuclein inclusions has been reported in greater proportions in GBA PD than non-GBA PD.[142]


#
#

Rare Causes and Risk Factors

Additional genes have been identified as possible causes or risk factors for PD and atypical parkinsonism. Analyses of ADPD families initially identified CHCHD2, TMEM230, and RIC3 as causative genes. Multiple mutations in CHCHD2 have been implicated among Chinese, Japanese, and European PD patients,[143] [144] [145] but not consistently supported in case-control studies[146] [147] [148]; recent meta-analyses suggest the P2L mutation is associated with increased PD risk among Asian populations.[149] [150] TMEM230 was reported in a North American ADPD family,[151] but has not been replicated in recent case-control studies and requires further validation.[152] [153] RIC3 was recently reported in an Indian ADPD family, but currently lacks validation.[154] Possible rare risk factors include SMPD1, which, in a biallelic state, causes Niemann-Pick disease type A or B, but is also associated with an increased risk of PD.[155] [156] [157] [158] Additional studies are required for DNAJC13, specific variants of which may be associated with increased PD risk.[159] Mutations in GCH1 are associated with dopa-responsive dystonia; in addition, reports including postmortem examination suggest an increased risk of parkinsonism with LB pathology among patients with dopa-responsive dystonia[160] in association with progressive supranuclear palsy-like pathology in one proven GCH1 patient.[161] 22q11.2 deletion causes CATCH22 syndrome, but is also associated with an increased risk for atypical parkinsonism[162] with supporting LB pathology on postmortem examination.[163]

Various genes have been associated with atypical parkinsonism. These include DNAJC6, associated with early-onset parkinsonism, mental retardation, hallucinations, pyramidal tract signs, and epilepsy[164] [165] [166] [167]; ATP13A2,[168] [169] associated with Kufor-Rakeb syndrome, characterized by early-onset, rapidly progressive parkinsonism with supranuclear gaze palsy, spasticity, and dementia; VPS13C,[170] associated with early-onset, rapidly progressive parkinsonism with early cognitive decline; SYNJ1,[171] associated with early-onset parkinsonism with dystonia as well as supranuclear gaze palsy, dementia, and seizures; FBXO7,[172] [173] associated with early-onset parkinsonism with pyramidal tract signs; PLA2G6,[174] associated with dystonia parkinsonism; and RAB39B,[175] [176] [177] associated with early-onset parkinsonism with childhood intellectual disability. Association with PD currently remains unclear as these genes are largely lacking postmortem examination to confirm or refute LB pathology.


#

Conclusion

One of the greatest advances in PD research in the past two decades is our better understanding of PD genetics. The wealth of genetic research helps confirm the clinical observation that PD is not a single disease with a single pathogenesis and natural course. However, much of the genetics of PD remains to be uncovered. Mutations in the genes outlined above and summarized in [Table 1] are present only in a small minority of people with PD; the cause of PD in most cases remains to be discovered. Also, despite extensive work, the mechanisms by which mutations in these genes cause PD remain largely unknown.

To date, most of the genetic research in PD has been observational. Studies have described the prevalence, phenotype, and clinical course of mutation carriers. Among the currently known genetic causes and risk factors, the clinical phenotype, including the manifestation of motor and nonmotor symptoms, and the rate of progression vary widely; these characteristics should be considerations in individual clinical management.

Our hope is that the genetic information may ultimately lead to new therapeutic interventions manipulating the metabolic pathways of the involved genes. In cases of loss of function (e.g., Parkin), interventions would aim to enhance the pathway and in cases of gain of function (e.g., LRRK2), to inhibit activity. Whether mutation carriers, or ideally the entire PD population, would benefit from such interventions remains to be explored.

Table 1

Major genetic causes and risk factors for Parkinson's disease: Ethnic distribution, phenotypic characteristics, neuropathological findings, and implications of heterozygosity and/or gene dosage

Gene

Populations

Phenotype

Pathology

Effect of heterozygosity

LRRK2

Varies by mutation: G2019S (N African Berber and AJ[21]); codon 1441 (Spanish Basque[9]); G2385R (Chinese, Japanese, Korean[12] [13] [14]); I2020T (Japanese[10])

Similar motor phenotype[19] and rate of progression as in iPD[25] with good levodopa response[26]

Fewer nonmotor manifestations than in iPD[18] [28] [29] [30] [33]

Substantia nigra and locus coeruleus degeneration with variable LB and tau pathology[40]

Causative with incomplete penetrance

SNCA

European[43] [46] [47] [48] [49]

Early-onset PD with more rapid progression than in iPD; good levodopa response but early fluctuations[50]; severe psychiatric features in gene duplication cases[51] [54]

Prominent cortical and subcortical LB pathology in triplication case[53]

Causative with high penetrance[52]; gene dosage effect noted[50]

VPS35

European,[57] [58] [60] Japanese[61]

Case reports only Tremor-predominant PD[61]

None available

Causative

Parkin

Worldwide

Early-onset PD with slow rate of progression

Mild cardinal features with some atypical features (dystonia, prominent freezing of gait), with sleep benefit on most symptoms[62] Less cognitive impairment than in iPD,[82] but more severe impulse control disorders[85]

Substantia nigra neuronal loss without prominent LB pathology[97] [98]

Possible risk factor with deletions, duplications, or point mutations in functionally critical domains[92] [93]

PINK1

Worldwide

Early-onset PD with slow rate of progression; good levodopa response[99]

Some atypical features: dystonia and sleep benefit[105]

Limited reports (n = 2) Substantia nigra neuronal loss with sparse or no LB pathology[112] [113]

Possible risk factor for late-onset PD[105] [110] [111]

DJ1

Dutch,[114] Italian[115]

Case reports only

Early-onset PD with reported blepharospasm; good levodopa response, but with motor fluctuations[115]

None available

Unknown

GBA

Worldwide: highest risk in AJ populations[129]

Similar motor phenotype as in iPD,[120] but with more rapid progression[133]

More severe nonmotor features than in iPD, particularly cognitive impairment[128] [135] [136] Earlier onset and more severe motor and nonmotor features in homozygotes/compound heterozygotes[139]

LB pathology; likely more cortical LBs than in noncarriers[140] [141]

Risk factor: penetrance increases with age[130] [131] and mutation severity[129] [132]; Gene dosage effect noted[139]

Abbreviations: AJ, Ashkenazi Jewish; iPD, idiopathic PD; LB, Lewy body.



#
#

Acknowledgments

Dr. Kim is supported by an Edmond J. Safra Fellowship in Movement Disorders through the Michael J. Fox Foundation for Parkinson's Research. The authors would like to thank Christopher Liong for reviewing and editing the manuscript.

  • References

  • 1 Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet 2009; 18 (R1): R48-R59
  • 2 Wszolek ZK, Pfeiffer B, Fulgham JR , et al. Western Nebraska family (family D) with autosomal dominant parkinsonism. Neurology 1995; 45 (3 Pt 1): 502-505
  • 3 Funayama M, Hasegawa K, Kowa H, Saito M, Tsuji S, Obata F. A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1. Ann Neurol 2002; 51 (3) 296-301
  • 4 Zimprich A, Biskup S, Leitner P , et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004; 44 (4) 601-607
  • 5 Paisán-Ruíz C, Jain S, Evans EW , et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron 2004; 44 (4) 595-600
  • 6 Rosenbusch KE, Kortholt A. Activation mechanism of LRRK2 and its cellular functions in Parkinson's disease. Parkinsons Dis 2016; 2016: 7351985
  • 7 West AB, Cookson MR. Identification of bona-fide LRRK2 kinase substrates. Mov Disord 2016; 31 (8) 1140-1141
  • 8 Pankratz N, Pauciulo MW, Elsaesser VE , et al; Parkinson Study Group-PROGENI Investigators. Mutations in LRRK2 other than G2019S are rare in a North American-based sample of familial Parkinson's disease. Mov Disord 2006; 21 (12) 2257-2260
  • 9 Gorostidi A, Ruiz-Martínez J, Lopez de Munain A, Alzualde A, Martí Massó JF. LRRK2 G2019S and R1441G mutations associated with Parkinson's disease are common in the Basque Country, but relative prevalence is determined by ethnicity. Neurogenetics 2009; 10 (2) 157-159
  • 10 Funayama M, Hasegawa K, Ohta E , et al. An LRRK2 mutation as a cause for the parkinsonism in the original PARK8 family. Ann Neurol 2005; 57 (6) 918-921
  • 11 Lu CS, Simons EJ, Wu-Chou YH , et al. The LRRK2 I2012T, G2019S, and I2020T mutations are rare in Taiwanese patients with sporadic Parkinson's disease. Parkinsonism Relat Disord 2005; 11 (8) 521-522
  • 12 Funayama M, Li Y, Tomiyama H , et al. Leucine-rich repeat kinase 2 G2385R variant is a risk factor for Parkinson disease in Asian population. Neuroreport 2007; 18 (3) 273-275
  • 13 Kim JM, Lee JY, Kim HJ , et al. The LRRK2 G2385R variant is a risk factor for sporadic Parkinson's disease in the Korean population. Parkinsonism Relat Disord 2010; 16 (2) 85-88
  • 14 Di Fonzo A, Wu-Chou YH, Lu CS , et al. A common missense variant in the LRRK2 gene, Gly2385Arg, associated with Parkinson's disease risk in Taiwan. Neurogenetics 2006; 7 (3) 133-138
  • 15 Fung HC, Chen CM, Hardy J, Singleton AB, Wu YR. A common genetic factor for Parkinson disease in ethnic Chinese population in Taiwan. BMC Neurol 2006; 6: 47
  • 16 Chan DK, Ng PW, Mok V , et al. LRRK2 Gly2385Arg mutation and clinical features in a Chinese population with early-onset Parkinson's disease compared to late-onset patients. J Neural Transm (Vienna) 2008; 115 (9) 1275-1277
  • 17 An XK, Peng R, Li T , et al. LRRK2 Gly2385Arg variant is a risk factor of Parkinson's disease among Han-Chinese from mainland China. Eur J Neurol 2008; 15 (3) 301-305
  • 18 Marras C, Alcalay RN, Caspell-Garcia C , et al; LRRK2 Cohort Consortium. Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson's disease. Mov Disord 2016; 31 (8) 1192-1202
  • 19 Aasly JO, Toft M, Fernandez-Mata I , et al. Clinical features of LRRK2-associated Parkinson's disease in central Norway. Ann Neurol 2005; 57 (5) 762-765
  • 20 Di Fonzo A, Rohé CF, Ferreira J , et al; Italian Parkinson Genetics Network. A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease. Lancet 2005; 365 (9457): 412-415
  • 21 Healy DG, Falchi M, O'Sullivan SS , et al; International LRRK2 Consortium. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study. Lancet Neurol 2008; 7 (7) 583-590
  • 22 Tan EK, Shen H, Tan LC , et al. The G2019S LRRK2 mutation is uncommon in an Asian cohort of Parkinson's disease patients. Neurosci Lett 2005; 384 (3) 327-329
  • 23 Marder K, Wang Y, Alcalay RN , et al; LRRK2 Ashkenazi Jewish Consortium. Age-specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish LRRK2 Consortium. Neurology 2015; 85 (1) 89-95
  • 24 Alcalay RN, Mejia-Santana H, Tang MX , et al. Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease. Arch Neurol 2009; 66 (12) 1517-1522
  • 25 Nabli F, Ben Sassi S, Amouri R, Duda JE, Farrer MJ, Hentati F. Motor phenotype of LRRK2-associated Parkinson's disease: a Tunisian longitudinal study. Mov Disord 2015; 30 (2) 253-258
  • 26 Deng H, Le W, Guo Y, Hunter CB, Xie W, Jankovic J. Genetic and clinical identification of Parkinson's disease patients with LRRK2 G2019S mutation. Ann Neurol 2005; 57 (6) 933-934
  • 27 Gilks WP, Abou-Sleiman PM, Gandhi S , et al. A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet 2005; 365 (9457): 415-416
  • 28 Ben Sassi S, Nabli F, Hentati E , et al. Cognitive dysfunction in Tunisian LRRK2 associated Parkinson's disease. Parkinsonism Relat Disord 2012; 18 (3) 243-246
  • 29 Alcalay RN, Mejia-Santana H, Mirelman A , et al; LRRK2 Ashkenazi Jewish Consortium. Neuropsychological performance in LRRK2 G2019S carriers with Parkinson's disease. Parkinsonism Relat Disord 2015; 21 (2) 106-110
  • 30 Marras C, Schüle B, Munhoz RP , et al. Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers. Neurology 2011; 77 (4) 325-333
  • 31 Saunders-Pullman R, Stanley K, San Luciano M , et al. Gender differences in the risk of familial parkinsonism: beyond LRRK2?. Neurosci Lett 2011; 496 (2) 125-128
  • 32 Saunders-Pullman R, Mirelman A, Wang C , et al. Olfactory identification in LRRK2 G2019S mutation carriers: a relevant marker?. Ann Clin Transl Neurol 2014; 1 (9) 670-678
  • 33 Saunders-Pullman R, Alcalay RN, Mirelman A , et al; AJ LRRK2 Consortium. REM sleep behavior disorder, as assessed by questionnaire, in G2019S LRRK2 mutation PD and carriers. Mov Disord 2015; 30 (13) 1834-1839
  • 34 Saunders-Pullman R, Barrett MJ, Stanley KM , et al. LRRK2 G2019S mutations are associated with an increased cancer risk in Parkinson disease. Mov Disord 2010; 25 (15) 2536-2541
  • 35 Inzelberg R, Cohen OS, Aharon-Peretz J , et al. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology 2012; 78 (11) 781-786
  • 36 Saunders-Pullman R, Stanley K, Wang C , et al. Olfactory dysfunction in LRRK2 G2019S mutation carriers. Neurology 2011; 77 (4) 319-324
  • 37 Mirelman A, Alcalay RN, Saunders-Pullman R , et al; LRRK2 AJ consortium. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov Disord 2015; 30 (7) 981-986
  • 38 Mirelman A, Bernad-Elazari H, Thaler A , et al. Arm swing as a potential new prodromal marker of Parkinson's disease. Mov Disord 2016; 31 (10) 1527-1534
  • 39 Shanker V, Groves M, Heiman G , et al. Mood and cognition in leucine-rich repeat kinase 2 G2019S Parkinson's disease. Mov Disord 2011; 26 (10) 1875-1880
  • 40 Poulopoulos M, Cortes E, Vonsattel JP , et al. Clinical and pathological characteristics of LRRK2 G2019S patients with PD. J Mol Neurosci 2012; 47 (1) 139-143
  • 41 Kalia LV, Lang AE, Hazrati LN , et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol 2015; 72 (1) 100-105
  • 42 Polymeropoulos MH, Higgins JJ, Golbe LI , et al. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science 1996; 274 (5290): 1197-1199
  • 43 Polymeropoulos MH, Lavedan C, Leroy E , et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997; 276 (5321): 2045-2047
  • 44 Chan P, Tanner CM, Jiang X, Langston JW. Failure to find the alpha-synuclein gene missense mutation (G209A) in 100 patients with younger onset Parkinson's disease. Neurology 1998; 50 (2) 513-514
  • 45 Scott WK, Yamaoka LH, Stajich JM , et al. The alpha-synuclein gene is not a major risk factor in familial Parkinson disease. Neurogenetics 1999; 2 (3) 191-192
  • 46 Zarranz JJ, Alegre J, Gómez-Esteban JC , et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004; 55 (2) 164-173
  • 47 Krüger R, Kuhn W, Müller T , et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet 1998; 18 (2) 106-108
  • 48 Proukakis C, Dudzik CG, Brier T , et al. A novel α-synuclein missense mutation in Parkinson disease. Neurology 2013; 80 (11) 1062-1064
  • 49 Lesage S, Anheim M, Letournel F , et al; French Parkinson's Disease Genetics Study Group. G51D α-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol 2013; 73 (4) 459-471
  • 50 Kasten M, Klein C. The many faces of alpha-synuclein mutations. Mov Disord 2013; 28 (6) 697-701
  • 51 Chartier-Harlin MC, Kachergus J, Roumier C , et al. Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 2004; 364 (9440): 1167-1169
  • 52 Papadimitriou D, Antonelou R, Miligkos M , et al. Motor and nonmotor features of carriers of the p.A53T alpha-synuclein mutation: a longitudinal study. Mov Disord 2016; 31 (8) 1226-1230
  • 53 Singleton AB, Farrer M, Johnson J , et al. alpha-Synuclein locus triplication causes Parkinson's disease. Science 2003; 302 (5646): 841
  • 54 Nishioka K, Ross OA, Ishii K , et al. Expanding the clinical phenotype of SNCA duplication carriers. Mov Disord 2009; 24 (12) 1811-1819
  • 55 Ahn TB, Kim SY, Kim JY , et al. alpha-Synuclein gene duplication is present in sporadic Parkinson disease. Neurology 2008; 70 (1) 43-49
  • 56 Ibáñez P, Bonnet AM, Débarges B , et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson's disease. Lancet 2004; 364 (9440): 1169-1171
  • 57 Vilariño-Güell C, Wider C, Ross OA , et al. VPS35 mutations in Parkinson disease. Am J Hum Genet 2011; 89 (1) 162-167
  • 58 Zimprich A, Benet-Pagès A, Struhal W , et al. A mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson disease. Am J Hum Genet 2011; 89 (1) 168-175
  • 59 Guella I, Soldà G, Cilia R , et al. The Asp620asn mutation in VPS35 is not a common cause of familial Parkinson's disease. Mov Disord 2012; 27 (6) 800-801
  • 60 Lesage S, Condroyer C, Klebe S , et al; French Parkinson's Disease Genetics Study Group. Identification of VPS35 mutations replicated in French families with Parkinson disease. Neurology 2012; 78 (18) 1449-1450
  • 61 Ando M, Funayama M, Li Y , et al. VPS35 mutation in Japanese patients with typical Parkinson's disease. Mov Disord 2012; 27 (11) 1413-1417
  • 62 Ishikawa A, Tsuji S. Clinical analysis of 17 patients in 12 Japanese families with autosomal-recessive type juvenile parkinsonism. Neurology 1996; 47 (1) 160-166
  • 63 Matsumine H, Saito M, Shimoda-Matsubayashi S , et al. Localization of a gene for an autosomal recessive form of juvenile Parkinsonism to chromosome 6q25.2-27. Am J Hum Genet 1997; 60 (3) 588-596
  • 64 Kitada T, Asakawa S, Hattori N , et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998; 392 (6676): 605-608
  • 65 Hattori N, Kitada T, Matsumine H , et al. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol 1998; 44 (6) 935-941
  • 66 Jeon BS, Kim JM, Lee DS, Hattori N, Mizuno Y. An apparently sporadic case with parkin gene mutation in a Korean woman. Arch Neurol 2001; 58 (6) 988-989
  • 67 Lücking CB, Abbas N, Dürr A , et al; The European Consortium on Genetic Susceptibility in Parkinson's Disease and the French Parkinson's Disease Genetics Study Group. Homozygous deletions in parkin gene in European and North African families with autosomal recessive juvenile parkinsonism. Lancet 1998; 352 (9137): 1355-1356
  • 68 Hedrich K, Marder K, Harris J , et al. Evaluation of 50 probands with early-onset Parkinson's disease for Parkin mutations. Neurology 2002; 58 (8) 1239-1246
  • 69 Marder KS, Tang MX, Mejia-Santana H , et al. Predictors of parkin mutations in early-onset Parkinson disease: the consortium on risk for early-onset Parkinson disease study. Arch Neurol 2010; 67 (6) 731-738
  • 70 Li X, Gehring K. Structural studies of parkin and sacsin: mitochondrial dynamics in neurodegenerative diseases. Mov Disord 2015; 30 (12) 1610-1619
  • 71 Rub C, Wilkening A, Voos W. Mitochondrial quality control by the Pink1/Parkin system. Cell Tissue Res 2017; 367 (1) 111-123
  • 72 Kilarski LL, Pearson JP, Newsway V , et al. Systematic review and UK-based study of PARK2 (parkin), PINK1, PARK7 (DJ-1) and LRRK2 in early-onset Parkinson's disease. Mov Disord 2012; 27 (12) 1522-1529
  • 73 Hedrich K, Eskelson C, Wilmot B , et al. Distribution, type, and origin of Parkin mutations: review and case studies. Mov Disord 2004; 19 (10) 1146-1157
  • 74 Klein C, Schumacher K, Jacobs H , et al. Association studies of Parkinson's disease and parkin polymorphisms. Ann Neurol 2000; 48 (1) 126-127
  • 75 Deng H, Le WD, Hunter CB , et al. Heterogeneous phenotype in a family with compound heterozygous parkin gene mutations. Arch Neurol 2006; 63 (2) 273-277
  • 76 Kann M, Hedrich K, Vieregge P , et al. The parkin gene is not involved in late-onset Parkinson's disease. Neurology 2002; 58 (5) 835 , author reply 835
  • 77 Oliveira SA, Scott WK, Martin ER , et al. Parkin mutations and susceptibility alleles in late-onset Parkinson's disease. Ann Neurol 2003; 53 (5) 624-629
  • 78 Klein C, Hedrich K, Wellenbrock C , et al. Frequency of parkin mutations in late-onset Parkinson's disease. Ann Neurol 2003; 54 (3) 415-416 , author reply 416–417
  • 79 Khan NL, Brooks DJ, Pavese N , et al. Progression of nigrostriatal dysfunction in a parkin kindred: an [18F]dopa PET and clinical study. Brain 2002; 125 (Pt 10): 2248-2256
  • 80 Lohmann E, Thobois S, Lesage S , et al; French Parkinson's Disease Genetics Study Group. A multidisciplinary study of patients with early-onset PD with and without parkin mutations. Neurology 2009; 72 (2) 110-116
  • 81 Caccappolo E, Alcalay RN, Mejia-Santana H , et al. Neuropsychological profile of Parkin mutation carriers with and without Parkinson disease: the CORE-PD Study. J Int Neuropsychol Soc 2011; 17 (1) 91-100
  • 82 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Cognitive and motor function in long-duration PARKIN-associated Parkinson disease. JAMA Neurol 2014; 71 (1) 62-67
  • 83 Khan NL, Katzenschlager R, Watt H , et al. Olfaction differentiates parkin disease from early-onset parkinsonism and Parkinson disease. Neurology 2004; 62 (7) 1224-1226
  • 84 Alcalay RN, Siderowf A, Ottman R , et al. Olfaction in Parkin heterozygotes and compound heterozygotes: the CORE-PD study. Neurology 2011; 76 (4) 319-326
  • 85 Morgante F, Fasano A, Ginevrino M , et al. Impulsive-compulsive behaviors in parkin-associated Parkinson disease. Neurology 2016; 87 (14) 1436-1441
  • 86 Cesari R, Martin ES, Calin GA , et al. Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27. Proc Natl Acad Sci U S A 2003; 100 (10) 5956-5961
  • 87 Fujiwara M, Marusawa H, Wang HQ , et al. Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene 2008; 27 (46) 6002-6011
  • 88 Poulogiannis G, McIntyre RE, Dimitriadi M , et al. PARK2 deletions occur frequently in sporadic colorectal cancer and accelerate adenoma development in Apc mutant mice. Proc Natl Acad Sci U S A 2010; 107 (34) 15145-15150
  • 89 Veeriah S, Taylor BS, Meng S , et al. Somatic mutations of the Parkinson's disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet 2010; 42 (1) 77-82
  • 90 Alcalay RN, Clark LN, Marder KS, Bradley WE. Lack of association between cancer history and PARKIN genotype: a family based study in PARKIN/Parkinson's families. Genes Chromosomes Cancer 2012; 51 (12) 1109-1113
  • 91 Kay DM, Moran D, Moses L , et al. Heterozygous parkin point mutations are as common in control subjects as in Parkinson's patients. Ann Neurol 2007; 61 (1) 47-54
  • 92 Kay DM, Stevens CF, Hamza TH , et al. A comprehensive analysis of deletions, multiplications, and copy number variations in PARK2. Neurology 2010; 75 (13) 1189-1194
  • 93 Pankratz N, Kissell DK, Pauciulo MW , et al; Parkinson Study Group-PROGENI Investigators. Parkin dosage mutations have greater pathogenicity in familial PD than simple sequence mutations. Neurology 2009; 73 (4) 279-286
  • 94 Romito LM, Contarino MF, Ghezzi D, Franzini A, Garavaglia B, Albanese A. High frequency stimulation of the subthalamic nucleus is efficacious in Parkin disease. J Neurol 2005; 252 (2) 208-211
  • 95 Moro E, Volkmann J, König IR , et al. Bilateral subthalamic stimulation in Parkin and PINK1 parkinsonism. Neurology 2008; 70 (14) 1186-1191
  • 96 Angeli A, Mencacci NE, Duran R , et al. Genotype and phenotype in Parkinson's disease: lessons in heterogeneity from deep brain stimulation. Mov Disord 2013; 28 (10) 1370-1375
  • 97 Pramstaller PP, Schlossmacher MG, Jacques TS , et al. Lewy body Parkinson's disease in a large pedigree with 77 Parkin mutation carriers. Ann Neurol 2005; 58 (3) 411-422
  • 98 Doherty KM, Silveira-Moriyama L, Parkkinen L , et al. Parkin disease: a clinicopathologic entity?. JAMA Neurol 2013; 70 (5) 571-579
  • 99 Valente EM, Bentivoglio AR, Dixon PH , et al. Localization of a novel locus for autosomal recessive early-onset parkinsonism, PARK6, on human chromosome 1p35-p36. Am J Hum Genet 2001; 68 (4) 895-900
  • 100 Valente EM, Abou-Sleiman PM, Caputo V , et al. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science 2004; 304 (5674): 1158-1160
  • 101 Valente EM, Salvi S, Ialongo T , et al. PINK1 mutations are associated with sporadic early-onset parkinsonism. Ann Neurol 2004; 56 (3) 336-341
  • 102 Rogaeva E, Johnson J, Lang AE , et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch Neurol 2004; 61 (12) 1898-1904
  • 103 Ibáñez P, Lesage S, Lohmann E , et al; French Parkinson's Disease Genetics Study Group. Mutational analysis of the PINK1 gene in early-onset parkinsonism in Europe and North Africa. Brain 2006; 129 (Pt 3): 686-694
  • 104 Hatano Y, Sato K, Elibol B , et al. PARK6-linked autosomal recessive early-onset parkinsonism in Asian populations. Neurology 2004; 63 (8) 1482-1485
  • 105 Bonifati V, Rohé CF, Breedveld GJ , et al; Italian Parkinson Genetics Network. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes. Neurology 2005; 65 (1) 87-95
  • 106 Truban D, Hou X, Caulfield TR, Fiesel FC, Springer W. PINK1, Parkin, and mitochondrial quality control: what can we learn about Parkinson's disease pathobiology?. J Parkinsons Dis 2017; 7 (1) 13-29
  • 107 Bentivoglio AR, Cortelli P, Valente EM , et al. Phenotypic characterisation of autosomal recessive PARK6-linked parkinsonism in three unrelated Italian families. Mov Disord 2001; 16 (6) 999-1006
  • 108 Ephraty L, Porat O, Israeli D , et al. Neuropsychiatric and cognitive features in autosomal-recessive early parkinsonism due to PINK1 mutations. Mov Disord 2007; 22 (4) 566-569
  • 109 Ferraris A, Ialongo T, Passali GC , et al. Olfactory dysfunction in parkinsonism caused by PINK1 mutations. Mov Disord 2009; 24 (16) 2350-2357
  • 110 Abou-Sleiman PM, Muqit MM, McDonald NQ , et al. A heterozygous effect for PINK1 mutations in Parkinson's disease?. Ann Neurol 2006; 60 (4) 414-419
  • 111 Kasten M, Weichert C, Lohmann K, Klein C. Clinical and demographic characteristics of PINK1 mutation carriers--a meta-analysis. Mov Disord 2010; 25 (7) 952-954
  • 112 Samaranch L, Lorenzo-Betancor O, Arbelo JM , et al. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain 2010; 133 (Pt 4): 1128-1142
  • 113 Takanashi M, Li Y, Hattori N. Absence of Lewy pathology associated with PINK1 homozygous mutation. Neurology 2016; 86 (23) 2212-2213
  • 114 van Duijn CM, Dekker MC, Bonifati V , et al. Park7, a novel locus for autosomal recessive early-onset parkinsonism, on chromosome 1p36. Am J Hum Genet 2001; 69 (3) 629-634
  • 115 Bonifati V, Rizzu P, van Baren MJ , et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003; 299 (5604): 256-259
  • 116 Sironi F, Primignani P, Ricca S , et al. DJ1 analysis in a large cohort of Italian early onset Parkinson disease patients. Neurosci Lett 2013; 557 (Pt B): 165-170
  • 117 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Frequency of known mutations in early-onset Parkinson disease: implication for genetic counseling: the consortium on risk for early onset Parkinson disease study. Arch Neurol 2010; 67 (9) 1116-1122
  • 118 Maraganore DM, Wilkes K, Lesnick TG , et al. A limited role for DJ1 in Parkinson disease susceptibility. Neurology 2004; 63 (3) 550-553
  • 119 Neudorfer O, Giladi N, Elstein D , et al. Occurrence of Parkinson's syndrome in type I Gaucher disease. QJM 1996; 89 (9) 691-694
  • 120 Aharon-Peretz J, Badarny S, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson disease: phenotype-genotype correlation. Neurology 2005; 65 (9) 1460-1461
  • 121 Beavan MS, Schapira AH. Glucocerebrosidase mutations and the pathogenesis of Parkinson disease. Ann Med 2013; 45 (8) 511-521
  • 122 Alcalay RN, Levy OA, Waters CC , et al. Glucocerebrosidase activity in Parkinson's disease with and without GBA mutations. Brain 2015; 138 (Pt 9): 2648-2658
  • 123 Huang CL, Wu-Chou YH, Lai SC , et al. Contribution of glucocerebrosidase mutation in a large cohort of sporadic Parkinson's disease in Taiwan. Eur J Neurol 2011; 18 (10) 1227-1232
  • 124 Wu YR, Chen CM, Chao CY , et al. Glucocerebrosidase gene mutation is a risk factor for early onset of Parkinson disease among Taiwanese. J Neurol Neurosurg Psychiatry 2007; 78 (9) 977-979
  • 125 Lesage S, Anheim M, Condroyer C , et al; French Parkinson's Disease Genetics Study Group. Large-scale screening of the Gaucher's disease-related glucocerebrosidase gene in Europeans with Parkinson's disease. Hum Mol Genet 2011; 20 (1) 202-210
  • 126 Neumann J, Bras J, Deas E , et al. Glucocerebrosidase mutations in clinical and pathologically proven Parkinson's disease. Brain 2009; 132 (Pt 7): 1783-1794
  • 127 Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson's disease in Ashkenazi Jews. N Engl J Med 2004; 351 (19) 1972-1977
  • 128 Sidransky E, Nalls MA, Aasly JO , et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease. N Engl J Med 2009; 361 (17) 1651-1661
  • 129 Gan-Or Z, Giladi N, Rozovski U , et al. Genotype-phenotype correlations between GBA mutations and Parkinson disease risk and onset. Neurology 2008; 70 (24) 2277-2283
  • 130 Anheim M, Elbaz A, Lesage S , et al; French Parkinson Disease Genetic Group. Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology 2012; 78 (6) 417-420
  • 131 Alcalay RN, Dinur T, Quinn T , et al. Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes. JAMA Neurol 2014; 71 (6) 752-757
  • 132 Gan-Or Z, Amshalom I, Kilarski LL , et al. Differential effects of severe vs mild GBA mutations on Parkinson disease. Neurology 2015; 84 (9) 880-887
  • 133 Brockmann K, Srulijes K, Pflederer S , et al. GBA-associated Parkinson's disease: reduced survival and more rapid progression in a prospective longitudinal study. Mov Disord 2015; 30 (3) 407-411
  • 134 Oeda T, Umemura A, Mori Y , et al. Impact of glucocerebrosidase mutations on motor and nonmotor complications in Parkinson's disease. Neurobiol Aging 2015; 36 (12) 3306-3313
  • 135 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Cognitive performance of GBA mutation carriers with early-onset PD: the CORE-PD study. Neurology 2012; 78 (18) 1434-1440
  • 136 Brockmann K, Srulijes K, Hauser AK , et al. GBA-associated PD presents with nonmotor characteristics. Neurology 2011; 77 (3) 276-280
  • 137 Gan-Or Z, Mirelman A, Postuma RB , et al. GBA mutations are associated with rapid eye movement sleep behavior disorder. Ann Clin Transl Neurol 2015; 2 (9) 941-945
  • 138 Cilia R, Tunesi S, Marotta G , et al. Survival and dementia in GBA-associated Parkinson's disease: the mutation matters. Ann Neurol 2016; 80 (5) 662-673
  • 139 Thaler A, Gurevich T, Bar Shira A , et al. A “dose” effect of mutations in the GBA gene on Parkinson's disease phenotype. Parkinsonism Relat Disord 2016; S1353-8020(16)30495-3
  • 140 Clark LN, Kartsaklis LA, Wolf Gilbert R , et al. Association of glucocerebrosidase mutations with dementia with Lewy bodies. Arch Neurol 2009; 66 (5) 578-583
  • 141 Parkkinen L, Neumann J, O'Sullivan SS , et al. Glucocerebrosidase mutations do not cause increased Lewy body pathology in Parkinson's disease. Mol Genet Metab 2011; 103 (4) 410-412
  • 142 Goker-Alpan O, Stubblefield BK, Giasson BI, Sidransky E. Glucocerebrosidase is present in α-synuclein inclusions in Lewy body disorders. Acta Neuropathol 2010; 120 (5) 641-649
  • 143 Funayama M, Ohe K, Amo T , et al. CHCHD2 mutations in autosomal dominant late-onset Parkinson's disease: a genome-wide linkage and sequencing study. Lancet Neurol 2015; 14 (3) 274-282
  • 144 Foo JN, Liu J, Tan EK. CHCHD2 and Parkinson's disease. Lancet Neurol 2015; 14 (7) 681-682
  • 145 Koschmidder E, Weissbach A, Brüggemann N, Kasten M, Klein C, Lohmann K. A nonsense mutation in CHCHD2 in a patient with Parkinson disease. Neurology 2016; 86 (6) 577-579
  • 146 Gagliardi M, Iannello G, Colica C, Annesi G, Quattrone A. Analysis of CHCHD2 gene in familial Parkinson's disease from Calabria. Neurobiol Aging 2017; 50: 169.e5-169.e6
  • 147 Tejera-Parrado C, Jesús S, Huertas-Fernández I , et al. Genetic analysis of CHCHD2 in a southern Spanish population. Neurobiol Aging 2017; 50: 169.e1-169.e2
  • 148 Fan TS, Lin HI, Lin CH, Wu RM. Lack of CHCHD2 mutations in Parkinson's disease in a Taiwanese population. Neurobiol Aging 2016; 38: 218.e1-218.e2
  • 149 Li NN, Wang L, Tan EK , et al. Genetic analysis of CHCHD2 gene in Chinese Parkinson's disease. Am J Med Genet B Neuropsychiatr Genet 2016; 171 (8) 1148-1152
  • 150 Yang X, Zhao Q, An R , et al. Mutational scanning of the CHCHD2 gene in Han Chinese patients with Parkinson's disease and meta-analysis of the literature. Parkinsonism Relat Disord 2016; 29: 42-46
  • 151 Deng HX, Shi Y, Yang Y , et al. Identification of TMEM230 mutations in familial Parkinson's disease. Nat Genet 2016; 48 (7) 733-739
  • 152 Yan W, Tang B, Zhou X , et al. TMEM230 mutation analysis in Parkinson's disease in a Chinese population. Neurobiol Aging 2017; 49: 219.e1-219.e3
  • 153 Giri A, Mok KY, Jansen I , et al; International Parkinson's Disease Consortium (IPDGC). Lack of evidence for a role of genetic variation in TMEM230 in the risk for Parkinson's disease in the Caucasian population. Neurobiol Aging 2017; 50: 167.e11-167.e13
  • 154 Sudhaman S, Muthane UB, Behari M, Govindappa ST, Juyal RC, Thelma BK. Evidence of mutations in RIC3 acetylcholine receptor chaperone as a novel cause of autosomal-dominant Parkinson's disease with non-motor phenotypes. J Med Genet 2016; 53 (8) 559-566
  • 155 Gan-Or Z, Ozelius LJ, Bar-Shira A , et al. The p.L302P mutation in the lysosomal enzyme gene SMPD1 is a risk factor for Parkinson disease. Neurology 2013; 80 (17) 1606-1610
  • 156 Foo JN, Liany H, Bei JX , et al. Rare lysosomal enzyme gene SMPD1 variant (p.R591C) associates with Parkinson's disease. Neurobiol Aging 2013; 34 (12) 2890.e13-2890.e15
  • 157 Dagan E, Adir V, Schlesinger I , et al. SMPD1 mutations and Parkinson disease. Parkinsonism Relat Disord 2015; 21 (10) 1296-1297
  • 158 Gan-Or Z, Orr-Urtreger A, Alcalay RN, Bressman S, Giladi N, Rouleau GA. The emerging role of SMPD1 mutations in Parkinson's disease: implications for future studies. Parkinsonism Relat Disord 2015; 21 (10) 1294-1295
  • 159 Vilariño-Güell C, Rajput A, Milnerwood AJ , et al. DNAJC13 mutations in Parkinson disease. Hum Mol Genet 2014; 23 (7) 1794-1801
  • 160 Gibb WR, Narabayashi H, Yokochi M, Iizuka R, Lees AJ. New pathologic observations in juvenile onset parkinsonism with dystonia. Neurology 1991; 41 (6) 820-822
  • 161 Guella I, Sherman HE, Appel-Cresswell S, Rajput A, Rajput AH, Farrer MJ. Parkinsonism in GTP cyclohydrolase 1 mutation carriers. Brain 2015; 138 (Pt 5): e349
  • 162 Mok KY, Sheerin U, Simón-Sánchez J , et al; International Parkinson's Disease Genomics Consortium (IPDGC). Deletions at 22q11.2 in idiopathic Parkinson's disease: a combined analysis of genome-wide association data. Lancet Neurol 2016; 15 (6) 585-596
  • 163 Butcher NJ, Kiehl TR, Hazrati LN , et al. Association between early-onset Parkinson disease and 22q11.2 deletion syndrome: identification of a novel genetic form of Parkinson disease and its clinical implications. JAMA Neurol 2013; 70 (11) 1359-1366
  • 164 Köroğlu Ç, Baysal L, Cetinkaya M, Karasoy H, Tolun A. DNAJC6 is responsible for juvenile parkinsonism with phenotypic variability. Parkinsonism Relat Disord 2013; 19 (3) 320-324
  • 165 Edvardson S, Cinnamon Y, Ta-Shma A , et al. A deleterious mutation in DNAJC6 encoding the neuronal-specific clathrin-uncoating co-chaperone auxilin, is associated with juvenile parkinsonism. PLoS One 2012; 7 (5) e36458
  • 166 Elsayed LE, Drouet V, Usenko T , et al. A novel nonsense mutation in DNAJC6 expands the phenotype of autosomal-recessive juvenile-onset Parkinson's disease. Ann Neurol 2016; 79 (2) 335-337
  • 167 Olgiati S, Quadri M, Fang M , et al; International Parkinsonism Genetics Network. DNAJC6 mutations associated with early-onset Parkinson's disease. Ann Neurol 2016; 79 (2) 244-256
  • 168 Najim al-Din AS, Wriekat A, Mubaidin A, Dasouki M, Hiari M. Pallido-pyramidal degeneration, supranuclear upgaze paresis and dementia: Kufor-Rakeb syndrome. Acta Neurol Scand 1994; 89 (5) 347-352
  • 169 Park JS, Blair NF, Sue CM. The role of ATP13A2 in Parkinson's disease: clinical phenotypes and molecular mechanisms. Mov Disord 2015; 30 (6) 770-779
  • 170 Lesage S, Drouet V, Majounie E , et al; French Parkinson's Disease Genetics Study (PDG); International Parkinson's Disease Genomics Consortium (IPDGC). Loss of VPS13C function in autosomal-recessive parkinsonism causes mitochondrial dysfunction and increases PINK1/Parkin-dependent mitophagy. Am J Hum Genet 2016; 98 (3) 500-513
  • 171 Quadri M, Fang M, Picillo M , et al; International Parkinsonism Genetics Network. Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset Parkinsonism. Hum Mutat 2013; 34 (9) 1208-1215
  • 172 Di Fonzo A, Dekker MC, Montagna P , et al. FBXO7 mutations cause autosomal recessive, early-onset parkinsonian-pyramidal syndrome. Neurology 2009; 72 (3) 240-245
  • 173 Lohmann E, Coquel AS, Honoré A , et al. A new F-box protein 7 gene mutation causing typical Parkinson's disease. Mov Disord 2015; 30 (8) 1130-1133
  • 174 Malaguti MC, Melzi V, Di Giacopo R , et al. A novel homozygous PLA2G6 mutation causes dystonia-parkinsonism. Parkinsonism Relat Disord 2015; 21 (3) 337-339
  • 175 Wilson GR, Sim JC, McLean C , et al. Mutations in RAB39B cause X-linked intellectual disability and early-onset Parkinson disease with α-synuclein pathology. Am J Hum Genet 2014; 95 (6) 729-735
  • 176 Mata IF, Jang Y, Kim CH , et al. The RAB39B p.G192R mutation causes X-linked dominant Parkinson's disease. Mol Neurodegener 2015; 10: 50
  • 177 Lesage S, Bras J, Cormier-Dequaire F , et al; French Parkinson's Disease Genetics Study Group (PDG) and the International Parkinson's Disease Genomics Consortium (IPDGC). Loss-of-function mutations in RAB39B are associated with typical early-onset Parkinson disease. Neurol Genet 2015; 1 (1) e9

Address for correspondence

Roy N. Alcalay, MD, MS
Department of Neurology, Columbia University Medical Center
710 W 168th St, 3rd floor, New York
NY 10032

  • References

  • 1 Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet 2009; 18 (R1): R48-R59
  • 2 Wszolek ZK, Pfeiffer B, Fulgham JR , et al. Western Nebraska family (family D) with autosomal dominant parkinsonism. Neurology 1995; 45 (3 Pt 1): 502-505
  • 3 Funayama M, Hasegawa K, Kowa H, Saito M, Tsuji S, Obata F. A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1. Ann Neurol 2002; 51 (3) 296-301
  • 4 Zimprich A, Biskup S, Leitner P , et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004; 44 (4) 601-607
  • 5 Paisán-Ruíz C, Jain S, Evans EW , et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron 2004; 44 (4) 595-600
  • 6 Rosenbusch KE, Kortholt A. Activation mechanism of LRRK2 and its cellular functions in Parkinson's disease. Parkinsons Dis 2016; 2016: 7351985
  • 7 West AB, Cookson MR. Identification of bona-fide LRRK2 kinase substrates. Mov Disord 2016; 31 (8) 1140-1141
  • 8 Pankratz N, Pauciulo MW, Elsaesser VE , et al; Parkinson Study Group-PROGENI Investigators. Mutations in LRRK2 other than G2019S are rare in a North American-based sample of familial Parkinson's disease. Mov Disord 2006; 21 (12) 2257-2260
  • 9 Gorostidi A, Ruiz-Martínez J, Lopez de Munain A, Alzualde A, Martí Massó JF. LRRK2 G2019S and R1441G mutations associated with Parkinson's disease are common in the Basque Country, but relative prevalence is determined by ethnicity. Neurogenetics 2009; 10 (2) 157-159
  • 10 Funayama M, Hasegawa K, Ohta E , et al. An LRRK2 mutation as a cause for the parkinsonism in the original PARK8 family. Ann Neurol 2005; 57 (6) 918-921
  • 11 Lu CS, Simons EJ, Wu-Chou YH , et al. The LRRK2 I2012T, G2019S, and I2020T mutations are rare in Taiwanese patients with sporadic Parkinson's disease. Parkinsonism Relat Disord 2005; 11 (8) 521-522
  • 12 Funayama M, Li Y, Tomiyama H , et al. Leucine-rich repeat kinase 2 G2385R variant is a risk factor for Parkinson disease in Asian population. Neuroreport 2007; 18 (3) 273-275
  • 13 Kim JM, Lee JY, Kim HJ , et al. The LRRK2 G2385R variant is a risk factor for sporadic Parkinson's disease in the Korean population. Parkinsonism Relat Disord 2010; 16 (2) 85-88
  • 14 Di Fonzo A, Wu-Chou YH, Lu CS , et al. A common missense variant in the LRRK2 gene, Gly2385Arg, associated with Parkinson's disease risk in Taiwan. Neurogenetics 2006; 7 (3) 133-138
  • 15 Fung HC, Chen CM, Hardy J, Singleton AB, Wu YR. A common genetic factor for Parkinson disease in ethnic Chinese population in Taiwan. BMC Neurol 2006; 6: 47
  • 16 Chan DK, Ng PW, Mok V , et al. LRRK2 Gly2385Arg mutation and clinical features in a Chinese population with early-onset Parkinson's disease compared to late-onset patients. J Neural Transm (Vienna) 2008; 115 (9) 1275-1277
  • 17 An XK, Peng R, Li T , et al. LRRK2 Gly2385Arg variant is a risk factor of Parkinson's disease among Han-Chinese from mainland China. Eur J Neurol 2008; 15 (3) 301-305
  • 18 Marras C, Alcalay RN, Caspell-Garcia C , et al; LRRK2 Cohort Consortium. Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson's disease. Mov Disord 2016; 31 (8) 1192-1202
  • 19 Aasly JO, Toft M, Fernandez-Mata I , et al. Clinical features of LRRK2-associated Parkinson's disease in central Norway. Ann Neurol 2005; 57 (5) 762-765
  • 20 Di Fonzo A, Rohé CF, Ferreira J , et al; Italian Parkinson Genetics Network. A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease. Lancet 2005; 365 (9457): 412-415
  • 21 Healy DG, Falchi M, O'Sullivan SS , et al; International LRRK2 Consortium. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study. Lancet Neurol 2008; 7 (7) 583-590
  • 22 Tan EK, Shen H, Tan LC , et al. The G2019S LRRK2 mutation is uncommon in an Asian cohort of Parkinson's disease patients. Neurosci Lett 2005; 384 (3) 327-329
  • 23 Marder K, Wang Y, Alcalay RN , et al; LRRK2 Ashkenazi Jewish Consortium. Age-specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish LRRK2 Consortium. Neurology 2015; 85 (1) 89-95
  • 24 Alcalay RN, Mejia-Santana H, Tang MX , et al. Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease. Arch Neurol 2009; 66 (12) 1517-1522
  • 25 Nabli F, Ben Sassi S, Amouri R, Duda JE, Farrer MJ, Hentati F. Motor phenotype of LRRK2-associated Parkinson's disease: a Tunisian longitudinal study. Mov Disord 2015; 30 (2) 253-258
  • 26 Deng H, Le W, Guo Y, Hunter CB, Xie W, Jankovic J. Genetic and clinical identification of Parkinson's disease patients with LRRK2 G2019S mutation. Ann Neurol 2005; 57 (6) 933-934
  • 27 Gilks WP, Abou-Sleiman PM, Gandhi S , et al. A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet 2005; 365 (9457): 415-416
  • 28 Ben Sassi S, Nabli F, Hentati E , et al. Cognitive dysfunction in Tunisian LRRK2 associated Parkinson's disease. Parkinsonism Relat Disord 2012; 18 (3) 243-246
  • 29 Alcalay RN, Mejia-Santana H, Mirelman A , et al; LRRK2 Ashkenazi Jewish Consortium. Neuropsychological performance in LRRK2 G2019S carriers with Parkinson's disease. Parkinsonism Relat Disord 2015; 21 (2) 106-110
  • 30 Marras C, Schüle B, Munhoz RP , et al. Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers. Neurology 2011; 77 (4) 325-333
  • 31 Saunders-Pullman R, Stanley K, San Luciano M , et al. Gender differences in the risk of familial parkinsonism: beyond LRRK2?. Neurosci Lett 2011; 496 (2) 125-128
  • 32 Saunders-Pullman R, Mirelman A, Wang C , et al. Olfactory identification in LRRK2 G2019S mutation carriers: a relevant marker?. Ann Clin Transl Neurol 2014; 1 (9) 670-678
  • 33 Saunders-Pullman R, Alcalay RN, Mirelman A , et al; AJ LRRK2 Consortium. REM sleep behavior disorder, as assessed by questionnaire, in G2019S LRRK2 mutation PD and carriers. Mov Disord 2015; 30 (13) 1834-1839
  • 34 Saunders-Pullman R, Barrett MJ, Stanley KM , et al. LRRK2 G2019S mutations are associated with an increased cancer risk in Parkinson disease. Mov Disord 2010; 25 (15) 2536-2541
  • 35 Inzelberg R, Cohen OS, Aharon-Peretz J , et al. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology 2012; 78 (11) 781-786
  • 36 Saunders-Pullman R, Stanley K, Wang C , et al. Olfactory dysfunction in LRRK2 G2019S mutation carriers. Neurology 2011; 77 (4) 319-324
  • 37 Mirelman A, Alcalay RN, Saunders-Pullman R , et al; LRRK2 AJ consortium. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov Disord 2015; 30 (7) 981-986
  • 38 Mirelman A, Bernad-Elazari H, Thaler A , et al. Arm swing as a potential new prodromal marker of Parkinson's disease. Mov Disord 2016; 31 (10) 1527-1534
  • 39 Shanker V, Groves M, Heiman G , et al. Mood and cognition in leucine-rich repeat kinase 2 G2019S Parkinson's disease. Mov Disord 2011; 26 (10) 1875-1880
  • 40 Poulopoulos M, Cortes E, Vonsattel JP , et al. Clinical and pathological characteristics of LRRK2 G2019S patients with PD. J Mol Neurosci 2012; 47 (1) 139-143
  • 41 Kalia LV, Lang AE, Hazrati LN , et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol 2015; 72 (1) 100-105
  • 42 Polymeropoulos MH, Higgins JJ, Golbe LI , et al. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science 1996; 274 (5290): 1197-1199
  • 43 Polymeropoulos MH, Lavedan C, Leroy E , et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997; 276 (5321): 2045-2047
  • 44 Chan P, Tanner CM, Jiang X, Langston JW. Failure to find the alpha-synuclein gene missense mutation (G209A) in 100 patients with younger onset Parkinson's disease. Neurology 1998; 50 (2) 513-514
  • 45 Scott WK, Yamaoka LH, Stajich JM , et al. The alpha-synuclein gene is not a major risk factor in familial Parkinson disease. Neurogenetics 1999; 2 (3) 191-192
  • 46 Zarranz JJ, Alegre J, Gómez-Esteban JC , et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004; 55 (2) 164-173
  • 47 Krüger R, Kuhn W, Müller T , et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet 1998; 18 (2) 106-108
  • 48 Proukakis C, Dudzik CG, Brier T , et al. A novel α-synuclein missense mutation in Parkinson disease. Neurology 2013; 80 (11) 1062-1064
  • 49 Lesage S, Anheim M, Letournel F , et al; French Parkinson's Disease Genetics Study Group. G51D α-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol 2013; 73 (4) 459-471
  • 50 Kasten M, Klein C. The many faces of alpha-synuclein mutations. Mov Disord 2013; 28 (6) 697-701
  • 51 Chartier-Harlin MC, Kachergus J, Roumier C , et al. Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 2004; 364 (9440): 1167-1169
  • 52 Papadimitriou D, Antonelou R, Miligkos M , et al. Motor and nonmotor features of carriers of the p.A53T alpha-synuclein mutation: a longitudinal study. Mov Disord 2016; 31 (8) 1226-1230
  • 53 Singleton AB, Farrer M, Johnson J , et al. alpha-Synuclein locus triplication causes Parkinson's disease. Science 2003; 302 (5646): 841
  • 54 Nishioka K, Ross OA, Ishii K , et al. Expanding the clinical phenotype of SNCA duplication carriers. Mov Disord 2009; 24 (12) 1811-1819
  • 55 Ahn TB, Kim SY, Kim JY , et al. alpha-Synuclein gene duplication is present in sporadic Parkinson disease. Neurology 2008; 70 (1) 43-49
  • 56 Ibáñez P, Bonnet AM, Débarges B , et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson's disease. Lancet 2004; 364 (9440): 1169-1171
  • 57 Vilariño-Güell C, Wider C, Ross OA , et al. VPS35 mutations in Parkinson disease. Am J Hum Genet 2011; 89 (1) 162-167
  • 58 Zimprich A, Benet-Pagès A, Struhal W , et al. A mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson disease. Am J Hum Genet 2011; 89 (1) 168-175
  • 59 Guella I, Soldà G, Cilia R , et al. The Asp620asn mutation in VPS35 is not a common cause of familial Parkinson's disease. Mov Disord 2012; 27 (6) 800-801
  • 60 Lesage S, Condroyer C, Klebe S , et al; French Parkinson's Disease Genetics Study Group. Identification of VPS35 mutations replicated in French families with Parkinson disease. Neurology 2012; 78 (18) 1449-1450
  • 61 Ando M, Funayama M, Li Y , et al. VPS35 mutation in Japanese patients with typical Parkinson's disease. Mov Disord 2012; 27 (11) 1413-1417
  • 62 Ishikawa A, Tsuji S. Clinical analysis of 17 patients in 12 Japanese families with autosomal-recessive type juvenile parkinsonism. Neurology 1996; 47 (1) 160-166
  • 63 Matsumine H, Saito M, Shimoda-Matsubayashi S , et al. Localization of a gene for an autosomal recessive form of juvenile Parkinsonism to chromosome 6q25.2-27. Am J Hum Genet 1997; 60 (3) 588-596
  • 64 Kitada T, Asakawa S, Hattori N , et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998; 392 (6676): 605-608
  • 65 Hattori N, Kitada T, Matsumine H , et al. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol 1998; 44 (6) 935-941
  • 66 Jeon BS, Kim JM, Lee DS, Hattori N, Mizuno Y. An apparently sporadic case with parkin gene mutation in a Korean woman. Arch Neurol 2001; 58 (6) 988-989
  • 67 Lücking CB, Abbas N, Dürr A , et al; The European Consortium on Genetic Susceptibility in Parkinson's Disease and the French Parkinson's Disease Genetics Study Group. Homozygous deletions in parkin gene in European and North African families with autosomal recessive juvenile parkinsonism. Lancet 1998; 352 (9137): 1355-1356
  • 68 Hedrich K, Marder K, Harris J , et al. Evaluation of 50 probands with early-onset Parkinson's disease for Parkin mutations. Neurology 2002; 58 (8) 1239-1246
  • 69 Marder KS, Tang MX, Mejia-Santana H , et al. Predictors of parkin mutations in early-onset Parkinson disease: the consortium on risk for early-onset Parkinson disease study. Arch Neurol 2010; 67 (6) 731-738
  • 70 Li X, Gehring K. Structural studies of parkin and sacsin: mitochondrial dynamics in neurodegenerative diseases. Mov Disord 2015; 30 (12) 1610-1619
  • 71 Rub C, Wilkening A, Voos W. Mitochondrial quality control by the Pink1/Parkin system. Cell Tissue Res 2017; 367 (1) 111-123
  • 72 Kilarski LL, Pearson JP, Newsway V , et al. Systematic review and UK-based study of PARK2 (parkin), PINK1, PARK7 (DJ-1) and LRRK2 in early-onset Parkinson's disease. Mov Disord 2012; 27 (12) 1522-1529
  • 73 Hedrich K, Eskelson C, Wilmot B , et al. Distribution, type, and origin of Parkin mutations: review and case studies. Mov Disord 2004; 19 (10) 1146-1157
  • 74 Klein C, Schumacher K, Jacobs H , et al. Association studies of Parkinson's disease and parkin polymorphisms. Ann Neurol 2000; 48 (1) 126-127
  • 75 Deng H, Le WD, Hunter CB , et al. Heterogeneous phenotype in a family with compound heterozygous parkin gene mutations. Arch Neurol 2006; 63 (2) 273-277
  • 76 Kann M, Hedrich K, Vieregge P , et al. The parkin gene is not involved in late-onset Parkinson's disease. Neurology 2002; 58 (5) 835 , author reply 835
  • 77 Oliveira SA, Scott WK, Martin ER , et al. Parkin mutations and susceptibility alleles in late-onset Parkinson's disease. Ann Neurol 2003; 53 (5) 624-629
  • 78 Klein C, Hedrich K, Wellenbrock C , et al. Frequency of parkin mutations in late-onset Parkinson's disease. Ann Neurol 2003; 54 (3) 415-416 , author reply 416–417
  • 79 Khan NL, Brooks DJ, Pavese N , et al. Progression of nigrostriatal dysfunction in a parkin kindred: an [18F]dopa PET and clinical study. Brain 2002; 125 (Pt 10): 2248-2256
  • 80 Lohmann E, Thobois S, Lesage S , et al; French Parkinson's Disease Genetics Study Group. A multidisciplinary study of patients with early-onset PD with and without parkin mutations. Neurology 2009; 72 (2) 110-116
  • 81 Caccappolo E, Alcalay RN, Mejia-Santana H , et al. Neuropsychological profile of Parkin mutation carriers with and without Parkinson disease: the CORE-PD Study. J Int Neuropsychol Soc 2011; 17 (1) 91-100
  • 82 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Cognitive and motor function in long-duration PARKIN-associated Parkinson disease. JAMA Neurol 2014; 71 (1) 62-67
  • 83 Khan NL, Katzenschlager R, Watt H , et al. Olfaction differentiates parkin disease from early-onset parkinsonism and Parkinson disease. Neurology 2004; 62 (7) 1224-1226
  • 84 Alcalay RN, Siderowf A, Ottman R , et al. Olfaction in Parkin heterozygotes and compound heterozygotes: the CORE-PD study. Neurology 2011; 76 (4) 319-326
  • 85 Morgante F, Fasano A, Ginevrino M , et al. Impulsive-compulsive behaviors in parkin-associated Parkinson disease. Neurology 2016; 87 (14) 1436-1441
  • 86 Cesari R, Martin ES, Calin GA , et al. Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27. Proc Natl Acad Sci U S A 2003; 100 (10) 5956-5961
  • 87 Fujiwara M, Marusawa H, Wang HQ , et al. Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene 2008; 27 (46) 6002-6011
  • 88 Poulogiannis G, McIntyre RE, Dimitriadi M , et al. PARK2 deletions occur frequently in sporadic colorectal cancer and accelerate adenoma development in Apc mutant mice. Proc Natl Acad Sci U S A 2010; 107 (34) 15145-15150
  • 89 Veeriah S, Taylor BS, Meng S , et al. Somatic mutations of the Parkinson's disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet 2010; 42 (1) 77-82
  • 90 Alcalay RN, Clark LN, Marder KS, Bradley WE. Lack of association between cancer history and PARKIN genotype: a family based study in PARKIN/Parkinson's families. Genes Chromosomes Cancer 2012; 51 (12) 1109-1113
  • 91 Kay DM, Moran D, Moses L , et al. Heterozygous parkin point mutations are as common in control subjects as in Parkinson's patients. Ann Neurol 2007; 61 (1) 47-54
  • 92 Kay DM, Stevens CF, Hamza TH , et al. A comprehensive analysis of deletions, multiplications, and copy number variations in PARK2. Neurology 2010; 75 (13) 1189-1194
  • 93 Pankratz N, Kissell DK, Pauciulo MW , et al; Parkinson Study Group-PROGENI Investigators. Parkin dosage mutations have greater pathogenicity in familial PD than simple sequence mutations. Neurology 2009; 73 (4) 279-286
  • 94 Romito LM, Contarino MF, Ghezzi D, Franzini A, Garavaglia B, Albanese A. High frequency stimulation of the subthalamic nucleus is efficacious in Parkin disease. J Neurol 2005; 252 (2) 208-211
  • 95 Moro E, Volkmann J, König IR , et al. Bilateral subthalamic stimulation in Parkin and PINK1 parkinsonism. Neurology 2008; 70 (14) 1186-1191
  • 96 Angeli A, Mencacci NE, Duran R , et al. Genotype and phenotype in Parkinson's disease: lessons in heterogeneity from deep brain stimulation. Mov Disord 2013; 28 (10) 1370-1375
  • 97 Pramstaller PP, Schlossmacher MG, Jacques TS , et al. Lewy body Parkinson's disease in a large pedigree with 77 Parkin mutation carriers. Ann Neurol 2005; 58 (3) 411-422
  • 98 Doherty KM, Silveira-Moriyama L, Parkkinen L , et al. Parkin disease: a clinicopathologic entity?. JAMA Neurol 2013; 70 (5) 571-579
  • 99 Valente EM, Bentivoglio AR, Dixon PH , et al. Localization of a novel locus for autosomal recessive early-onset parkinsonism, PARK6, on human chromosome 1p35-p36. Am J Hum Genet 2001; 68 (4) 895-900
  • 100 Valente EM, Abou-Sleiman PM, Caputo V , et al. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science 2004; 304 (5674): 1158-1160
  • 101 Valente EM, Salvi S, Ialongo T , et al. PINK1 mutations are associated with sporadic early-onset parkinsonism. Ann Neurol 2004; 56 (3) 336-341
  • 102 Rogaeva E, Johnson J, Lang AE , et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch Neurol 2004; 61 (12) 1898-1904
  • 103 Ibáñez P, Lesage S, Lohmann E , et al; French Parkinson's Disease Genetics Study Group. Mutational analysis of the PINK1 gene in early-onset parkinsonism in Europe and North Africa. Brain 2006; 129 (Pt 3): 686-694
  • 104 Hatano Y, Sato K, Elibol B , et al. PARK6-linked autosomal recessive early-onset parkinsonism in Asian populations. Neurology 2004; 63 (8) 1482-1485
  • 105 Bonifati V, Rohé CF, Breedveld GJ , et al; Italian Parkinson Genetics Network. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes. Neurology 2005; 65 (1) 87-95
  • 106 Truban D, Hou X, Caulfield TR, Fiesel FC, Springer W. PINK1, Parkin, and mitochondrial quality control: what can we learn about Parkinson's disease pathobiology?. J Parkinsons Dis 2017; 7 (1) 13-29
  • 107 Bentivoglio AR, Cortelli P, Valente EM , et al. Phenotypic characterisation of autosomal recessive PARK6-linked parkinsonism in three unrelated Italian families. Mov Disord 2001; 16 (6) 999-1006
  • 108 Ephraty L, Porat O, Israeli D , et al. Neuropsychiatric and cognitive features in autosomal-recessive early parkinsonism due to PINK1 mutations. Mov Disord 2007; 22 (4) 566-569
  • 109 Ferraris A, Ialongo T, Passali GC , et al. Olfactory dysfunction in parkinsonism caused by PINK1 mutations. Mov Disord 2009; 24 (16) 2350-2357
  • 110 Abou-Sleiman PM, Muqit MM, McDonald NQ , et al. A heterozygous effect for PINK1 mutations in Parkinson's disease?. Ann Neurol 2006; 60 (4) 414-419
  • 111 Kasten M, Weichert C, Lohmann K, Klein C. Clinical and demographic characteristics of PINK1 mutation carriers--a meta-analysis. Mov Disord 2010; 25 (7) 952-954
  • 112 Samaranch L, Lorenzo-Betancor O, Arbelo JM , et al. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain 2010; 133 (Pt 4): 1128-1142
  • 113 Takanashi M, Li Y, Hattori N. Absence of Lewy pathology associated with PINK1 homozygous mutation. Neurology 2016; 86 (23) 2212-2213
  • 114 van Duijn CM, Dekker MC, Bonifati V , et al. Park7, a novel locus for autosomal recessive early-onset parkinsonism, on chromosome 1p36. Am J Hum Genet 2001; 69 (3) 629-634
  • 115 Bonifati V, Rizzu P, van Baren MJ , et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003; 299 (5604): 256-259
  • 116 Sironi F, Primignani P, Ricca S , et al. DJ1 analysis in a large cohort of Italian early onset Parkinson disease patients. Neurosci Lett 2013; 557 (Pt B): 165-170
  • 117 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Frequency of known mutations in early-onset Parkinson disease: implication for genetic counseling: the consortium on risk for early onset Parkinson disease study. Arch Neurol 2010; 67 (9) 1116-1122
  • 118 Maraganore DM, Wilkes K, Lesnick TG , et al. A limited role for DJ1 in Parkinson disease susceptibility. Neurology 2004; 63 (3) 550-553
  • 119 Neudorfer O, Giladi N, Elstein D , et al. Occurrence of Parkinson's syndrome in type I Gaucher disease. QJM 1996; 89 (9) 691-694
  • 120 Aharon-Peretz J, Badarny S, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson disease: phenotype-genotype correlation. Neurology 2005; 65 (9) 1460-1461
  • 121 Beavan MS, Schapira AH. Glucocerebrosidase mutations and the pathogenesis of Parkinson disease. Ann Med 2013; 45 (8) 511-521
  • 122 Alcalay RN, Levy OA, Waters CC , et al. Glucocerebrosidase activity in Parkinson's disease with and without GBA mutations. Brain 2015; 138 (Pt 9): 2648-2658
  • 123 Huang CL, Wu-Chou YH, Lai SC , et al. Contribution of glucocerebrosidase mutation in a large cohort of sporadic Parkinson's disease in Taiwan. Eur J Neurol 2011; 18 (10) 1227-1232
  • 124 Wu YR, Chen CM, Chao CY , et al. Glucocerebrosidase gene mutation is a risk factor for early onset of Parkinson disease among Taiwanese. J Neurol Neurosurg Psychiatry 2007; 78 (9) 977-979
  • 125 Lesage S, Anheim M, Condroyer C , et al; French Parkinson's Disease Genetics Study Group. Large-scale screening of the Gaucher's disease-related glucocerebrosidase gene in Europeans with Parkinson's disease. Hum Mol Genet 2011; 20 (1) 202-210
  • 126 Neumann J, Bras J, Deas E , et al. Glucocerebrosidase mutations in clinical and pathologically proven Parkinson's disease. Brain 2009; 132 (Pt 7): 1783-1794
  • 127 Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson's disease in Ashkenazi Jews. N Engl J Med 2004; 351 (19) 1972-1977
  • 128 Sidransky E, Nalls MA, Aasly JO , et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease. N Engl J Med 2009; 361 (17) 1651-1661
  • 129 Gan-Or Z, Giladi N, Rozovski U , et al. Genotype-phenotype correlations between GBA mutations and Parkinson disease risk and onset. Neurology 2008; 70 (24) 2277-2283
  • 130 Anheim M, Elbaz A, Lesage S , et al; French Parkinson Disease Genetic Group. Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology 2012; 78 (6) 417-420
  • 131 Alcalay RN, Dinur T, Quinn T , et al. Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes. JAMA Neurol 2014; 71 (6) 752-757
  • 132 Gan-Or Z, Amshalom I, Kilarski LL , et al. Differential effects of severe vs mild GBA mutations on Parkinson disease. Neurology 2015; 84 (9) 880-887
  • 133 Brockmann K, Srulijes K, Pflederer S , et al. GBA-associated Parkinson's disease: reduced survival and more rapid progression in a prospective longitudinal study. Mov Disord 2015; 30 (3) 407-411
  • 134 Oeda T, Umemura A, Mori Y , et al. Impact of glucocerebrosidase mutations on motor and nonmotor complications in Parkinson's disease. Neurobiol Aging 2015; 36 (12) 3306-3313
  • 135 Alcalay RN, Caccappolo E, Mejia-Santana H , et al. Cognitive performance of GBA mutation carriers with early-onset PD: the CORE-PD study. Neurology 2012; 78 (18) 1434-1440
  • 136 Brockmann K, Srulijes K, Hauser AK , et al. GBA-associated PD presents with nonmotor characteristics. Neurology 2011; 77 (3) 276-280
  • 137 Gan-Or Z, Mirelman A, Postuma RB , et al. GBA mutations are associated with rapid eye movement sleep behavior disorder. Ann Clin Transl Neurol 2015; 2 (9) 941-945
  • 138 Cilia R, Tunesi S, Marotta G , et al. Survival and dementia in GBA-associated Parkinson's disease: the mutation matters. Ann Neurol 2016; 80 (5) 662-673
  • 139 Thaler A, Gurevich T, Bar Shira A , et al. A “dose” effect of mutations in the GBA gene on Parkinson's disease phenotype. Parkinsonism Relat Disord 2016; S1353-8020(16)30495-3
  • 140 Clark LN, Kartsaklis LA, Wolf Gilbert R , et al. Association of glucocerebrosidase mutations with dementia with Lewy bodies. Arch Neurol 2009; 66 (5) 578-583
  • 141 Parkkinen L, Neumann J, O'Sullivan SS , et al. Glucocerebrosidase mutations do not cause increased Lewy body pathology in Parkinson's disease. Mol Genet Metab 2011; 103 (4) 410-412
  • 142 Goker-Alpan O, Stubblefield BK, Giasson BI, Sidransky E. Glucocerebrosidase is present in α-synuclein inclusions in Lewy body disorders. Acta Neuropathol 2010; 120 (5) 641-649
  • 143 Funayama M, Ohe K, Amo T , et al. CHCHD2 mutations in autosomal dominant late-onset Parkinson's disease: a genome-wide linkage and sequencing study. Lancet Neurol 2015; 14 (3) 274-282
  • 144 Foo JN, Liu J, Tan EK. CHCHD2 and Parkinson's disease. Lancet Neurol 2015; 14 (7) 681-682
  • 145 Koschmidder E, Weissbach A, Brüggemann N, Kasten M, Klein C, Lohmann K. A nonsense mutation in CHCHD2 in a patient with Parkinson disease. Neurology 2016; 86 (6) 577-579
  • 146 Gagliardi M, Iannello G, Colica C, Annesi G, Quattrone A. Analysis of CHCHD2 gene in familial Parkinson's disease from Calabria. Neurobiol Aging 2017; 50: 169.e5-169.e6
  • 147 Tejera-Parrado C, Jesús S, Huertas-Fernández I , et al. Genetic analysis of CHCHD2 in a southern Spanish population. Neurobiol Aging 2017; 50: 169.e1-169.e2
  • 148 Fan TS, Lin HI, Lin CH, Wu RM. Lack of CHCHD2 mutations in Parkinson's disease in a Taiwanese population. Neurobiol Aging 2016; 38: 218.e1-218.e2
  • 149 Li NN, Wang L, Tan EK , et al. Genetic analysis of CHCHD2 gene in Chinese Parkinson's disease. Am J Med Genet B Neuropsychiatr Genet 2016; 171 (8) 1148-1152
  • 150 Yang X, Zhao Q, An R , et al. Mutational scanning of the CHCHD2 gene in Han Chinese patients with Parkinson's disease and meta-analysis of the literature. Parkinsonism Relat Disord 2016; 29: 42-46
  • 151 Deng HX, Shi Y, Yang Y , et al. Identification of TMEM230 mutations in familial Parkinson's disease. Nat Genet 2016; 48 (7) 733-739
  • 152 Yan W, Tang B, Zhou X , et al. TMEM230 mutation analysis in Parkinson's disease in a Chinese population. Neurobiol Aging 2017; 49: 219.e1-219.e3
  • 153 Giri A, Mok KY, Jansen I , et al; International Parkinson's Disease Consortium (IPDGC). Lack of evidence for a role of genetic variation in TMEM230 in the risk for Parkinson's disease in the Caucasian population. Neurobiol Aging 2017; 50: 167.e11-167.e13
  • 154 Sudhaman S, Muthane UB, Behari M, Govindappa ST, Juyal RC, Thelma BK. Evidence of mutations in RIC3 acetylcholine receptor chaperone as a novel cause of autosomal-dominant Parkinson's disease with non-motor phenotypes. J Med Genet 2016; 53 (8) 559-566
  • 155 Gan-Or Z, Ozelius LJ, Bar-Shira A , et al. The p.L302P mutation in the lysosomal enzyme gene SMPD1 is a risk factor for Parkinson disease. Neurology 2013; 80 (17) 1606-1610
  • 156 Foo JN, Liany H, Bei JX , et al. Rare lysosomal enzyme gene SMPD1 variant (p.R591C) associates with Parkinson's disease. Neurobiol Aging 2013; 34 (12) 2890.e13-2890.e15
  • 157 Dagan E, Adir V, Schlesinger I , et al. SMPD1 mutations and Parkinson disease. Parkinsonism Relat Disord 2015; 21 (10) 1296-1297
  • 158 Gan-Or Z, Orr-Urtreger A, Alcalay RN, Bressman S, Giladi N, Rouleau GA. The emerging role of SMPD1 mutations in Parkinson's disease: implications for future studies. Parkinsonism Relat Disord 2015; 21 (10) 1294-1295
  • 159 Vilariño-Güell C, Rajput A, Milnerwood AJ , et al. DNAJC13 mutations in Parkinson disease. Hum Mol Genet 2014; 23 (7) 1794-1801
  • 160 Gibb WR, Narabayashi H, Yokochi M, Iizuka R, Lees AJ. New pathologic observations in juvenile onset parkinsonism with dystonia. Neurology 1991; 41 (6) 820-822
  • 161 Guella I, Sherman HE, Appel-Cresswell S, Rajput A, Rajput AH, Farrer MJ. Parkinsonism in GTP cyclohydrolase 1 mutation carriers. Brain 2015; 138 (Pt 5): e349
  • 162 Mok KY, Sheerin U, Simón-Sánchez J , et al; International Parkinson's Disease Genomics Consortium (IPDGC). Deletions at 22q11.2 in idiopathic Parkinson's disease: a combined analysis of genome-wide association data. Lancet Neurol 2016; 15 (6) 585-596
  • 163 Butcher NJ, Kiehl TR, Hazrati LN , et al. Association between early-onset Parkinson disease and 22q11.2 deletion syndrome: identification of a novel genetic form of Parkinson disease and its clinical implications. JAMA Neurol 2013; 70 (11) 1359-1366
  • 164 Köroğlu Ç, Baysal L, Cetinkaya M, Karasoy H, Tolun A. DNAJC6 is responsible for juvenile parkinsonism with phenotypic variability. Parkinsonism Relat Disord 2013; 19 (3) 320-324
  • 165 Edvardson S, Cinnamon Y, Ta-Shma A , et al. A deleterious mutation in DNAJC6 encoding the neuronal-specific clathrin-uncoating co-chaperone auxilin, is associated with juvenile parkinsonism. PLoS One 2012; 7 (5) e36458
  • 166 Elsayed LE, Drouet V, Usenko T , et al. A novel nonsense mutation in DNAJC6 expands the phenotype of autosomal-recessive juvenile-onset Parkinson's disease. Ann Neurol 2016; 79 (2) 335-337
  • 167 Olgiati S, Quadri M, Fang M , et al; International Parkinsonism Genetics Network. DNAJC6 mutations associated with early-onset Parkinson's disease. Ann Neurol 2016; 79 (2) 244-256
  • 168 Najim al-Din AS, Wriekat A, Mubaidin A, Dasouki M, Hiari M. Pallido-pyramidal degeneration, supranuclear upgaze paresis and dementia: Kufor-Rakeb syndrome. Acta Neurol Scand 1994; 89 (5) 347-352
  • 169 Park JS, Blair NF, Sue CM. The role of ATP13A2 in Parkinson's disease: clinical phenotypes and molecular mechanisms. Mov Disord 2015; 30 (6) 770-779
  • 170 Lesage S, Drouet V, Majounie E , et al; French Parkinson's Disease Genetics Study (PDG); International Parkinson's Disease Genomics Consortium (IPDGC). Loss of VPS13C function in autosomal-recessive parkinsonism causes mitochondrial dysfunction and increases PINK1/Parkin-dependent mitophagy. Am J Hum Genet 2016; 98 (3) 500-513
  • 171 Quadri M, Fang M, Picillo M , et al; International Parkinsonism Genetics Network. Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset Parkinsonism. Hum Mutat 2013; 34 (9) 1208-1215
  • 172 Di Fonzo A, Dekker MC, Montagna P , et al. FBXO7 mutations cause autosomal recessive, early-onset parkinsonian-pyramidal syndrome. Neurology 2009; 72 (3) 240-245
  • 173 Lohmann E, Coquel AS, Honoré A , et al. A new F-box protein 7 gene mutation causing typical Parkinson's disease. Mov Disord 2015; 30 (8) 1130-1133
  • 174 Malaguti MC, Melzi V, Di Giacopo R , et al. A novel homozygous PLA2G6 mutation causes dystonia-parkinsonism. Parkinsonism Relat Disord 2015; 21 (3) 337-339
  • 175 Wilson GR, Sim JC, McLean C , et al. Mutations in RAB39B cause X-linked intellectual disability and early-onset Parkinson disease with α-synuclein pathology. Am J Hum Genet 2014; 95 (6) 729-735
  • 176 Mata IF, Jang Y, Kim CH , et al. The RAB39B p.G192R mutation causes X-linked dominant Parkinson's disease. Mol Neurodegener 2015; 10: 50
  • 177 Lesage S, Bras J, Cormier-Dequaire F , et al; French Parkinson's Disease Genetics Study Group (PDG) and the International Parkinson's Disease Genomics Consortium (IPDGC). Loss-of-function mutations in RAB39B are associated with typical early-onset Parkinson disease. Neurol Genet 2015; 1 (1) e9