Neurosurg Focus 28 (1):E7, 2010

Genetic basis of Parkinson disease

Ge o r g i a Xiromerisiou , M.D.,1 Ef t h i m i o s Da r d i o t i s , M.D.,1,2 Va ï a Ts i m o u r t o u , M.D.,2 Pe r s a Ma r i a Ko u n t r a , M.D.,2 Konstantinos N. Pa t e r a k i s , M.D.,3 Ef t y c h i a Z. Ka p s a l a k i , M.D.,4 Ko s t a s N. Fo u n t a s , M.D.,3 a n d Ge o r g i o s M. Ha d j i g e o r g i o u , M.D.1,2 1Institute for Biomedical Technology (BIOMED), Centre for Research and Technology—Thessaly (CERETETH); 2Department of Neurology, Laboratory of Neurogenetics; 3Department of Neurosurgery; and 4Department of Diagnostic Radiology, University of Thessaly, University Hospital of Larissa, Greece

Over the past few years, considerable progress has been made in understanding the molecular mechanisms of Parkinson disease (PD). Mutations in certain are found to cause monogenic forms of the disorder, with au- tosomal dominant or autosomal recessive inheritance. These genes include alpha-synuclein, parkin, PINK1, DJ-1, LRRK2, and ATP13A2. The monogenic variants are important tools in identifying cellular pathways that shed light on the pathogenesis of this disease. Certain common genetic variants are also likely to modulate the risk of PD. In- ternational collaborative studies and meta-analyses have identified common variants as genetic susceptibility risk/ protective factors for sporadic PD. (DOI: 10.3171/2009.10.FOCUS09220)

Ke y Wo r d s • Parkinson disease • genetic association study • polymorphism • mutation

a r k i n s o n disease is the second most common neu- ance of certain clinical features, and also the disease pro- rodegenerative disorder after Alzheimer disease. gression. The average age at onset is between 60 and 80 In this paper, a concise summary of the main genes Pyears, and ~ 1% of the general population older than 65 responsible for monogenic forms of PD (Table 1), and is affected.12,89 some of the common genetic variants that may play a role Although PD appears to be sporadic in most cases, in the disease, is provided. linkage and positional cloning studies were instrumental in the identification of certain genes that cause familial forms of the disease that feature autosomal dominant Monogenic Forms of PD or autosomal recessive inheritance.12,89 Approximately Alpha-synuclein (SNCA-PARK1/PARK4) is cen- 5–10% of patients with the clinical picture of PD carry tral to the pathophysiological mechanisms of familial a mutation in one of the known genes that cause mono- and sporadic PD. This protein is the major component of genic forms of the disorder.12,89 There is some evidence Lewy bodies and Lewy neuritis in PD, and also in oth- that these genes may also play a role in the much more er synucleinopathies. Three missense mutations (A53T, common sporadic form of the disease. The biochemical A30P, E64 K), and duplications or triplications of the lo- function of these genes can underlie various pathoge- cus containing SNCA have been identified previously in netic pathways, such as oxidative stress, mitochondrial families with autosomal dominant PD.29,42,65,67,76,77,90,92 The dysfunction, disturbance in protein quality control, and phenotype of patients with SNCA point mutations is that altered kinase activity that may lead to PD. of levodopa-responsive parkinsonism in patients with a The vast majority of cases of this disorder are spo- relatively young age at onset, rapid progression, and high radic, resulting from complex interactions among genes, prevalence of dementia, psychiatric, and autonomic dis- and between genes and environmental factors. Genetic turbances. Patients with duplications resemble those with variations may be susceptibility factors for disease that idiopathic PD,10 and those with triplications have earlier also affect penetrance, age at onset, severity, the appear- onset, faster disease progression, severe dementia, and frequent dysautonomia.23,76 There is consensus that any Abbreviations used in this paper: DBS = deep brain stimulation; change in the levels of alpha-synuclein expression or the PD = Parkinson disease; SNP = single nucleotide polymorphism. presence of mutations in alpha-synuclein has a toxic ef-

Neurosurg Focus / Volume 28 / January 2010 1

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC G. Xiromerisiou et al.

TABLE 1: Genetic loci and genes associated with monogenic forms of PD*

Locus Gene Inheritance & Comments OMIM No. PARK1 SNCA AD; 1st PD gene identified 168601 PARK2 PRKN AR; most common cause of recessive juvenile PD 602544 PARK3 SPR? AD 602404 & 182125 PARK4 SNCA AD 168601 PARK5 UCHL1 AD 191342 PARK6 PINK1 AR; 2nd most common cause of recessive juvenile PD 605909 PARK7 DJ-1 AR 606324 PARK8 LRRK2 AD; most common cause of dominant PD 607060 PARK9 ATP13A2 AR; PD plus dementia & spasticity 606693 PARK10 unknown ? 606852 PARK11 GIGYF2? AD 607688 PARK12 unknown ? 300557 PARK13 Omi/HTRA2 ? 610297 PARK14 PLA2G6 AR; PD plus adult-onset dystonia 612953 PARK15 FBXO7 AR; PD plus dementia & spasticity 260300

* AD = autosomal dominant; AR = autosomal recessive; OMIM = Online Mendelian Inheritance in Man; ? = controversial or unidentified. fect on dopaminergic neurons.18 Alpha-synuclein mono- parkinsonism.81,82 This gene links PD to mitochondrial mers interact under certain circumstances to form proto- dysfunction and oxidative stress as it encodes a primar- fibrils or fibrillar β-pleated sheets.84,86,89 Toxicity caused ily mitochondrial protein kinase. This protein may exert a by protofibrils may involve the leakage of dopamine from protective effect on the cell.2,69 No pathological mechanism synaptic vesicles because of perforation of the vesicular has yet been reported in patients with PINK1 mutations. membranes by these protofibrils.29 This may account for Patients with mutations in this gene are characterized by the selective toxicity of alpha-synuclein in the dopamine- early onset of parkinsonism (between 32 and 48 years producing neurons of the substantia nigra.13,56,84 of age), with slow progression and sustained response to Parkin (PRKN, PARK2) was the first gene identi- levodopa. There are also some indications that they have a fied for an autosomal recessive form of PD.1,5,35,40,83 Par- higher prevalence of psychiatric disturbances.3,17 kin protein localizes, although not predominantly, to the The third for autosomal recessive juvenile par- synapse and associates with membranes. Its main func- kinsonism was mapped also to 1p36, and the tion is as an ubiquitin ligase in the cellular ubiquitina- gene was identified as the oncogene DJ-1 (PARK7).6 The tion protein degradation pathway. Severe and selective function of the DJ-1 protein is not entirely clear. The main degeneration in the substantia nigra pars compacta but hypothesis is that it acts as a sensor for oxidative stress, without Lewy bodies has been described, suggesting that providing neuroprotection in situations of increased de- the disease may differ in some important ways from typi- mand.7,93 Parkinson disease causing DJ-1 mutations is rare cal idiopathic PD.35,68,83,91 Parkin mutations turned out to and accounts for only ~ 1–2% of early-onset autosomal be a very common cause of parkinsonism. All types of recessive PD cases. The phenotype closely resembles that mutations; missense mutations, nonsense mutations, and found in patients with parkin and PINK1 mutations.6 The exonic rearrangements were identified. The vast major- associated pathological characteristics are still unknown, ity of patients with parkin mutations manifest the disease because no autopsies have been reported. before the age of 40 years, with slow progression, but with Mutations in LRRK2 (PARK8) have been found in a levodopa-associated fluctuations and dyskinesias occur- large number of patients with PD.63,94 More than 40 dif- ring early and frequently. Unusual features such as focal ferent variants, almost all missense mutations, have been dystonia, early postural instability, and autonomic failure reported.4,37 The G2019S mutation in particular was de- may also be present. tected in 5–6% of autosomal dominant familial PD cases Mutations in the PINK1 gene (PARK6) have also been and in 1–2% of sporadic cases.28,46,62,80 Specific popula- identified as a cause of autosomal recessive early-onset tions such as Ashkenazi Jews and North African Arabs

2 Neurosurg Focus / Volume 28 / January 2010

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC Genetic basis of Parkinson disease were found to have an even higher prevalence.45,62 The Neurosurgical Treatment in Familial PD LRRK2 is a large protein with multiple protein interac- tions and catalytic domains that have possible roles in The identification of inherited forms of PD has also intracellular signaling pathways.58,85 Some pathogenetic helped in developing a more appropriate application mutations seem to be associated with an increase of ki- of neurosurgical treatments of the disease. Deep brain nase activity, which appears to be necessary for neuro- stimulation provides symptomatic benefits to patients 32,39 with idiopathic PD in terms of both motor activity and toxicity in vitro. Although LRRK2 was not found to 72 interact with either alpha-synuclein or tau, the identifica- quality of life. In hereditary PD, DBS is an efficacious symptomatic treatment for patients with parkinsonism tion of Lewy body or neurofibrillary tangle pathological and mutation of the parkin and PINK1 genes.8,73 Pa- features in patients with LRRK2 mutations suggested a tients with parkin mutations are specifically expected to possible common role of LRRK2 in the processing of be very good candidates for DBS and to benefit more those two proteins.85 The phenotype of LRRK2 mutation and longer than other patients because of their younger carriers is that of idiopathic PD, although the whole clini- age at onset, lower daily doses of levodopa, and slower cal picture is more benign. disease progression.8,51,73 The response to DBS among patients carrying mutations of the LRRK2 gene is not Other Genes and Loci well established.46,75 There are some studies that show that the response to DBS was worse among patients with The ATP13A2 gene is mapped at the PARK9 locus the R1441G mutation in LRRK2 compared with patients and is responsible for Kufor-Rakeb disease, a recessive, with idiopathic PD. juvenile-onset, atypical Parkinsonism with pyramidal 71 Based on the aforementioned findings, we conclude degeneration and cognitive dysfunction. The ATP13A2 that the effectiveness of DBS in different genetic forms gene encodes a large lysosomal P-type adenosine triphos- of PD has not been studied adequately. The main reason phatase, which is involved in the lysosomal degradation for this is that patient selection is based predominantly pathway that clears SNCA aggregates. Lysosomal dys- on clinical criteria.9,52 A multidisciplinary approach in- function caused by mutations in this gene might contrib- 83 volving a neurosurgeon, a neurologist, and a neuropsy- ute to the pathogenesis of parkinsonism. chologist is important to determine the appropriate surgi- A heterozygous missense mutation in the ubiquitin cal candidate. The best prognostic indicator of a patient’s carboxy-terminal hydrolase L1 gene (UCHL1, PARK5), suitability for DBS surgery is his/her response to levodo- which is located on chromosome 4p, has been identified pa.9 The patient’s age also is another major factor deter- in a single affected family of German ancestry. However, mining how an individual will cope with the surgical 87 whether UCHL1 is really a PD gene is not clear yet. procedure and behave postoperatively.74 Based on these Mutations in the glucocerebrosidase gene (GBA) criteria, many patients with genetic forms of the disease (which has not been assigned a PARK locus yet) are the who have a sustained response to levodopa38 or atypical cause of a recessive lysosomal storage disorder—Gaucher symptoms are excluded from this procedure. As a result, disease. More than 200 mutations have been described in it is impossible, based solely on clinical characteristics of GBA. Phenotypes of Gaucher disease and PD do not over- monogenic PD forms, to study the effectiveness of DBS lap significantly, but the first indication for a relationship in these patients. Another reason for this is that most pa- between the two actually came from clinical descriptions. tients who have undergone DBS surgery are not screened Mutations in GBA have a conspicuously high prevalence for mutations in various genes responsible for monogenic in patients with PD.57 Between 2 and 4% of Caucasian forms of the disease. patients with PD have been found to have mutations in The detailed evaluation of these patients and the ge- GBA. Those variants are likely to act as risk factors rath- netic analysis based on criteria such as the family history, er than as high-penetrance disease genes.59 The nature of phenotype, age at onset, and response to levodopa are of the association between PD and Gaucher disease remains great importance. However, larger series of patients with elusive. However, the pathogenic mechanisms leading to mutations and longer follow-ups will be needed for evi- PD in carriers of mutant GBA may be related to the faulty dence of specific genotype-related differences. processing of toxic proteins, aggravated by the relative Another matter worth discussing is that there are decrease in GBA activity and accumulation of glucoce- also no studies showing the optimal DBS target based rebrosidase.50,61 Moreover, recent findings indicate that on genetics. Generally the literature demonstrates a trend Gaucher disease and PD share pathophysiological fea- that the subthalamic nucleus may be more efficient in tures.88 managing the symptoms of PD, based on institutional Other mendelian forms of PD remain to be identi- experience, surgical and programming management, fied. Either the causal genes at several loci have not yet lower current requirements, and significant reduction in been identified (PARK12), or the role of the candidate dopaminergic medication.38,41,66 For these reasons, all genes at these loci is still controversial (PARK3, PARK10, of the previous studies in patients with mutations have PARK13). Two novel genes, the FBXO7 (PARK15), a referred to the effectiveness of DBS of the subthalamic member of the F-box family of proteins active in the nucleus.30,51,75 Maybe the genetic forms of the disease with ubiquitin-proteasome protein degradation pathway,14 and the different neuropathological characteristics will enable the PLA2G6 gene (PARK14) on chromosome 22, have deciphering of the mechanisms of DBS on the basis of also been identified in families with atypical PD.33 the function and pathophysiological characteristics of the

Neurosurg Focus / Volume 28 / January 2010 3

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC G. Xiromerisiou et al. cortico-basal-thalamo-cortical loops when different DBS tion approach to unravel genetic susceptibility factors. targets are being applied to them. Genome-wide association studies of PD, which use haplo- type tagging strategies to study variation across the entire , provided little evidence until now about Genetic Susceptibility Factors in Sporadic PD genetic variants that influence the risk for disease. A Monogenic forms represent < 10% of cases of PD. 2-stage genome-wide association study with a 200-SNP Common PD, on the other hand, is thought to result from map, and a 1-stage study with more informative mark- complex interactions involving genetic and environmental ers found no positive associations.25,54 The most strongly risk factors. The discovery that 1-methyl-4-phenyl tetrahy- associated SNPs identified in the 2-stage study were not dropyridine, a contaminant of a synthetic opiate, can cause replicated in a subsequent association study with a large parkinsonism through its neurotoxic metabolite, 1-methyl- number of participants.16,54 However, the combination of 4-phenylpyridinium, stimulated interest in environmental genome-wide databases with meta-analytical techniques chemical exposures as risk factors for PD.44 Many studies can improve the detection of genetic variants with small have investigated the association between PD and pesticide effect sizes. The GAK-DGKQ region on use, and some, but not all, have found an association.22,70,95 has been identified by this strategy, albeit not replicated Use of well water, rural living, and agricultural employ- in a recent report.19,20 The genomic pathway approach that ment have also been implicated as risk factors, although combines SNPs with axon guidance pathway genes has studies have given conflicting results.43,95 also been applied to genome-wide association studies, On the other hand, the extent of the genetic compo- with one positive result,48 but again without replication.49 nent remains elusive. Common genetic variations (main- Genome-wide association studies require large sample se- ly SNPs) may be either susceptibility factors or disease ries and international collaborations, so probably we will modifiers, affecting penetrance, age at onset, severity, or have to wait for a few years to identify possible common disease progression. genetic risk variants and clearly understand their role in Genetic association studies that compare the fre- the disease.20,25,36,47,54 quency of putative risk alleles in cohorts of patients and controls are controversial because they have failed to reproduce the initial positive findings most of the time. Conclusions Almost 800 genetic association studies have been per- It is hoped that an understanding of the genetic basis formed so far on more than 500 genes regarding PD (see of PD will allow us to identify upstream key facts of the www.pdgene.org). The vast majority of genetic associa- pathogenesis and lead to new targeted therapeutic strate- tion studies have focused on candidate genes involved gies in the future. Large-scale multicenter collaborations, in detoxification of metabolites, dopamine metabolism, public availability of the International HapMap Project, mitochondrial function, and familial PD.78 Some of these and genome-wide association PD databases will hope- findings were exciting at the beginning because the en- fully arm researchers with information that could be used coded proteins of these genes appear to be closely linked for modifying the natural course of the disease. to the pathophysiological mechanisms of PD; however, none of these candidate gene variants have been consis- Disclosure tently replicated since then. Thus, theoretically attrac- tive, broad-based meta-analyses yielded no true common The authors report no conflict of interest concerning the mate- genetic risk variant. Potential biases include population rials or methods used in this study or the findings specified in this stratification, small sample size, misclassification, and -in paper. 36 Author contributions to the study and manuscript preparation appropriate statistical methods. include the following. Conception and design: E Dardiotis. Analysis Nevertheless, specific polymorphic variants have and interpretation of data: E Dardiotis, V Tsimourtou, PM Kountra, been validated as genetic susceptibility factors. The Rep KN Paterakis, EZ Kapsalaki. Study supervision: KN Fountas, G 1, a mixed nucleotide repeat in the promoter region of Xiromerisiou, GM Hadjigeorgiou. SNCA, has been confirmed as a risk factor.11,34,53,55,60,64 A polymorphism in microtubule-associated protein tau References has also been detected.26 The combination of risk geno- types in SNCA and microtubule-associated protein tau 1. Abbas N, Lücking CB, Ricard S, Dürr A, Bonifati V, De Mi- 27,31 chele G, et al: A wide variety of mutations in the parkin gene doubles the risk of PD. Two variants in the LRRK2 are responsible for autosomal recessive parkinsonism in Eu- gene, G2385R and R1628P, confer susceptibility to PD in rope. Hum Mol Genet 8:567–574, 1999 Asian populations.15,21,24,80 An S18Y variant of the UCHL1 2. Abou-Sleiman PM, Muqit MM, Wood NW: Expanding in- gene has been demonstrated to be protective against PD sights of mitochondrial dysfunction in Parkinson’s disease. in some association studies and meta-analyses.5,79 The Nat Rev Neurosci 7:207–219, 2006 number of polymorphisms that have been studied until 3. Albanese A, Valente EM, Romito LM, Bellacchio E, Elia AE, now is very large, but so far these are some of the main Dallapiccola B: The PINK1 phenotype can be indistinguish- able from idiopathic Parkinson disease. Neurology 64:1958– risk alleles for sporadic PD that seems to be robustly re- 1960, 2005 producible. 4. Berg D, Schweitzer K, Leitner P, Zimprich A, Lichtner P, Bel- With the completion of the human “HapMap” proj- credi P, et al: Type and frequency of mutations in the LRRK2 ect and the availability of the SNP databases, there is gene in familial and sporadic Parkinson’s disease*. Brain increasing interest in using the whole-genome associa- 128:3000–3011, 2005

4 Neurosurg Focus / Volume 28 / January 2010

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC Genetic basis of Parkinson disease

5. Betarbet R, Sherer TB, Greenamyre JT: Ubiquitin-proteasome disease: a population-based case-control study. Arch Neurol system and Parkinson’s diseases. Exp Neurol 191 (Suppl 62:91–95, 2005 1):S17–S27, 2005 23. Fuchs J, Nilsson C, Kachergus J, Munz M, Larsson EM, 6. Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld Schüle B, et al: Phenotypic variation in a large Swedish pedi- GJ, Krieger E, et al: Mutations in the DJ-1 gene associated gree due to SNCA duplication and triplication. Neurology with autosomal recessive early-onset parkinsonism. Science 68:916–922, 2007 299:256–259, 2003 24. Funayama M, Li Y, Tomiyama H, Yoshino H, Imamichi Y, 7. Canet-Avilés RM, Wilson MA, Miller DW, Ahmad R, McLen- Yamamoto M, et al: Leucine-rich repeat kinase 2 G2385R don C, Bandyopadhyay S, et al: The Parkinson’s disease pro- variant is a risk factor for Parkinson disease in Asian popula- tein DJ-1 is neuroprotective due to cysteine-sulfinic acid- tion. Neuroreport 18:273–275, 2007 driven mitochondrial localization. Proc Natl Acad Sci U S A 25. Fung HC, Scholz S, Matarin M, Simón-Sánchez J, Hernandez 101:9103–9108, 2004 D, Britton A, et al: Genome-wide genotyping in Parkinson’s 8. Capecci M, Passamonti L, Annesi F, Annesi G, Bellesi M, disease and neurologically normal controls: first stage analysis Candiano IC, et al: Chronic bilateral subthalamic deep brain and public release of data. Lancet Neurol 5:911–916, 2006 stimulation in a patient with homozygous deletion in the par- 26. Fung HC, Xiromerisiou G, Gibbs JR, Wu YR, Eerola J, Gour- kin gene. Mov Disord 19:1450–1452, 2004 bali V, et al: Association of tau haplotype-tagging polymor- 9. Charles PD, Van Blercom N, Krack P, Lee SL, Xie J, Besson phisms with Parkinson’s disease in diverse ethnic Parkinson’s G, et al: Predictors of effective bilateral subthalamic nucleus disease cohorts. Neurodegener Dis 3:327–333, 2006 stimulation for PD. Neurology 59:932–934, 2002 27. Galpern WR, Lang AE: Interface between tauopathies and sy- 10. Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, nucleinopathies: a tale of two proteins. Ann Neurol 59:449– Douay X, Lincoln S, et al: Alpha-synuclein locus duplication 458, 2006 as a cause of familial Parkinson’s disease. Lancet 364:1167– 28. Gilks WP, Abou-Sleiman PM, Gandhi S, Jain S, Singleton A, 1169, 2004 Lees AJ, et al: A common LRRK2 mutation in idiopathic Par- 11. Chiba-Falek O, Nussbaum RL: Effect of allelic variation at kinson’s disease. Lancet 365:415–416, 2005 the NACP-Rep1 repeat upstream of the alpha-synuclein gene 29. Goldberg MS, Lansbury PT Jr: Is there a cause-and-effect re- (SNCA) on transcription in a cell culture luciferase reporter lationship between alpha-synuclein fibrillization and Parkin- system. Hum Mol Genet 10:3101–3109, 2001 son’s disease? Nat Cell Biol 2:E115–E119, 2000 12. Cookson MR, Xiromerisiou G, Singleton A: How genetics 30. Gómez-Esteban JC, Lezcano E, Zarranz JJ, González C, research in Parkinson’s disease is enhancing understanding Bilbao G, Lambarri I, et al: Outcome of bilateral deep brain of the common idiopathic forms of the disease. Curr Opin subthalamic stimulation in patients carrying the R1441G mu- Neurol 18:706–711, 2005 tation in the LRRK2 dardarin gene. Neurosurgery 62:857– 13. Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer 863, 2008 D: Impaired degradation of mutant alpha-synuclein by chap- 31. Goris A, Williams-Gray CH, Clark GR, Foltynie T, Lewis SJ, erone-mediated autophagy. Science 305:1292–1295, 2004 Brown J, et al: Tau and alpha-synuclein in susceptibility to, 14. Di Fonzo A, Dekker MC, Montagna P, Baruzzi A, Yonova EH, and dementia in, Parkinson’s disease. Ann Neurol 62:145– Correia Guedes L, et al: FBXO7 mutations cause auto­somal re- 153, 2007 cessive, early-onset parkinsonian-pyramidal syndrome. Neu­ 32. Greggio E, Jain S, Kingsbury A, Bandopadhyay R, Lewis P, r o l ­o g y 72:240–245, 2009 Kaganovich A, et al: Kinase activity is required for the toxic 15. Di Fonzo A, Wu-Chou YH, Lu CS, van Doeselaar M, Simons effects of mutant LRRK2/dardarin. Neurobiol Dis 23:329– EJ, Rohé CF, et al: A common missense variant in the LRRK2 341, 2006 gene, Gly2385Arg, associated with Parkinson’s disease risk in 33. Gregory A, Westaway SK, Holm IE, Kotzbauer PT, Hogarth P, Taiwan. Neurogenetics 7:133–138, 2006 Sonek S, et al: Neurodegeneration associated with genetic de- 16. Elbaz A, Nelson LM, Payami H, Ioannidis JP, Fiske BK, fects in phospholipase A(2). Neurology 71:1402–1409, 2008 Annesi G, et al: Lack of replication of thirteen single-nucle- 34. Hadjigeorgiou GM, Xiromerisiou G, Gourbali V, Aggelakis otide polymorphisms implicated in Parkinson’s disease: a K, Scarmeas N, Papadimitriou A, et al: Association of alpha- large-scale international study. Lancet Neurol 5:917–923, synuclein Rep1 polymorphism and Parkinson’s disease: influ- 2006 ence of Rep1 on age at onset. Mov Disord 21:534–539, 2006 17. Ephraty L, Porat O, Israeli D, Cohen OS, Tunkel O, Yael S, 35. Hasegawa T, Treis A, Patenge N, Fiesel FC, Springer W, Kah- et al: Neuropsychiatric and cognitive features in autosomal- le PJ: Parkin protects against tyrosinase-mediated dopamine recessive early parkinsonism due to PINK1 mutations. Mov neurotoxicity by suppressing stress-activated protein kinase Disord 22:566–569, 2007 pathways. J Neurochem 105:1700–1715, 2008 18. Eriksen JL, Przedborski S, Petrucelli L: Gene dosage and 36. Hattersley AT, McCarthy MI: What makes a good genetic as- pathogenesis of Parkinson’s disease. Trends Mol Med 11:91– sociation study? Lancet 366:1315–1323, 2005 96, 2005 37. Healy DG, Falchi M, O’Sullivan SS, Bonifati V, Durr A, 19. Evangelou E, Maraganore DM, Annesi G, Brighina L, Brice Bressman S, et al: Phenotype, genotype, and worldwide ge- A, Elbaz A, et al: Non-replication of association for six poly- netic penetrance of LRRK2-associated Parkinson’s disease: a morphisms from meta-analysis of genome-wide association case-control study. Lancet Neurol 7:583–590, 2008 studies of Parkinson’s disease: large-scale collaborative study. 38. Herzog J, Volkmann J, Krack P, Kopper F, Pötter M, Lorenz D, Am J Med Genet B Neuropsychiatr Genet [epub ahead of et al: Two-year follow-up of subthalamic deep brain stimula- print], 2009 tion in Parkinson’s disease. Mov Disord 18:1332–1337, 2003 20. Evangelou E, Maraganore DM, Ioannidis JP: Meta-analysis in 39. Ito G, Okai T, Fujino G, Takeda K, Ichijo H, Katada T, et genome-wide association datasets: strategies and application al: GTP binding is essential to the protein kinase activity of in Parkinson disease. PLoS One 2:e196, 2007 LRRK2, a causative gene product for familial Parkinson’s 21. Farrer MJ, Stone JT, Lin CH, Dächsel JC, Hulihan MM, Hau- disease. Biochemistry 46:1380–1388, 2007 garvoll K, et al: Lrrk2 G2385R is an ancestral risk factor for 40. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Parkinson’s disease in Asia. Parkinsonism Relat Disord Minoshima S, et al: Mutations in the parkin gene cause auto- 13:89–92, 2007 somal recessive juvenile parkinsonism. Nature 392:605–608, 22. Firestone JA, Smith-Weller T, Franklin G, Swanson P, Long- 1998 streth WT Jr, Checkoway H: Pesticides and risk of Parkinson 41. Kleiner-Fisman G, Fisman DN, Sime E, Saint-Cyr JA, Lozano

Neurosurg Focus / Volume 28 / January 2010 5

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC G. Xiromerisiou et al.

AM, Lang AE: Long-term follow up of bilateral deep brain of nitric oxide synthase induction by 15-deoxyspergualin stimulation of the subthalamic nucleus in patients with ad- in a cultured macrophage cell line, J774A.1 [correction of vanced Parkinson disease. J Neurosurg 99:489–495, 2003 J744A.1] activated with IFN-gamma and LPS. J Antibiot 42. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, (Tokyo) 52:297–304, 1999 et al: Ala30Pro mutation in the gene encoding alpha-synuclein 62. Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, in Parkinson’s disease. Nat Genet 18:106–108, 1998 Deligtisch A, Tagliati M, et al: LRRK2 G2019S as a cause 43. Lai BC, Marion SA, Teschke K, Tsui JK: Occupational and of Parkinson’s disease in Ashkenazi Jews. N Engl J Med environmental risk factors for Parkinson’s disease. Parkin­ 354:424–425, 2006 sonism Relat Disord 8:297–309, 2002 63. Paisán-Ruíz C, Jain S, Evans EW, Gilks WP, Simón J, van der 44. Langston JW, Ballard P, Tetrud JW, Irwin I: Chronic Parkin- Brug M, et al: Cloning of the gene containing mutations that sonism in humans due to a product of meperidine-analog syn- cause PARK8-linked Parkinson’s disease. Neuron 44:595– thesis. Science 219:979–980, 1983 600, 2004 45. Lesage S, Dürr A, Tazir M, Lohmann E, Leutenegger AL, Ja- 64. Pals P, Lincoln S, Manning J, Heckman M, Skipper L, Huli- nin S, et al: LRRK2 G2019S as a cause of Parkinson’s disease han M, et al: α-Synuclein promoter confers susceptibility to in North African Arabs. N Engl J Med 354:422–423, 2006 Parkinson’s disease. Ann Neurol 56:591–595, 2004 46. Lesage S, Janin S, Lohmann E, Leutenegger AL, Leclere L, 65. Papadimitriou A, Veletza V, Hadjigeorgiou GM, Patrikiou A, Viallet F, et al: LRRK2 exon 41 mutations in sporadic Parkin- Hirano M, Anastasopoulos I: Mutated alpha-synuclein gene in son disease in Europeans. Arch Neurol 64:425–430, 2007 two Greek kindreds with familial PD: incomplete penetrance? 47. Lesnick TG, Papapetropoulos S, Mash DC, Ffrench-Mullen J, Neurology 52:651–654, 1999 Shehadeh L, de Andrade M, et al: A genomic pathway ap- 66. Peppe A, Pierantozzi M, Bassi A, Altibrandi MG, Brusa L, proach to a complex disease: axon guidance and Parkinson Stefani A, et al: Stimulation of the subthalamic nucleus com- disease. PLoS Genet 3:e98, 2007 pared with the globus pallidus internus in patients with Par- 48. Lesnick TG, Sorenson EJ, Ahlskog JE, Henley JR, Shehadeh kinson disease. J Neurosurg 101:195–200, 2004 L, Papapetropoulos S, et al: Beyond Parkinson disease: amyo- 67. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, trophic lateral sclerosis and the axon guidance pathway. PLoS Dutra A, et al: Mutation in the alpha-synuclein gene identified One 3:e1449, 2008 in families with Parkinson’s disease. Science 276:2045–2047, 49. Li Y, Rowland C, Xiromerisiou G, Lagier RJ, Schrodi SJ, Dra- 1997 diotis E, et al: Neither replication nor simulation supports a 68. Portman AT, Giladi N, Leenders KL, Maguire P, Veenma-van role for the axon guidance pathway in the genetics of Parkin- der Duin L, Swart J, et al: The nigrostriatal dopaminergic sys- son’s disease. PLoS One 3:e2707, 2008 tem in familial early onset parkinsonism with parkin muta- 50. Lloyd-Evans E, Pelled D, Riebeling C, Bodennec J, de-Morgan tions. Neurology 56:1759–1762, 2001 A, Waller H, et al: Glucosylceramide and glucosylsphingosine 69. Pridgeon JW, Olzmann JA, Chin LS, Li L: PINK1 protects modulate calcium mobilization from brain microsomes via against oxidative stress by phosphorylating mitochondrial different mechanisms. J Biol Chem 278:23594–23599, 2003 chaperone TRAP1. PLoS Biol 5:e172, 2007 51. Lohmann E, Welter ML, Fraix V, Krack P, Lesage S, Laine 70. Priyadarshi A, Khuder SA, Schaub EA, Shrivastava S: A me- S, et al: Are parkin patients particularly suited for deep-brain ta-analysis of Parkinson’s disease and exposure to pesticides. stimulation? Mov Disord 23:740–743, 2008 Neurotoxicology 21:435–440, 2000 52. Machado A, Rezai AR, Kopell BH, Gross RE, Sharan AD, 71. Ramirez A, Heimbach A, Gründemann J, Stiller B, Hampshire Benabid AL: Deep brain stimulation for Parkinson’s disease: D, Cid LP, et al: Hereditary parkinsonism with dementia is surgical technique and perioperative management. Mov Dis­ caused by mutations in ATP13A2, encoding a lysosomal type ord 21 Suppl 14:S247–S258, 2006 5 P-type ATPase. Nat Genet 38:1184–1191, 2006 53. Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioan- 72. Ricciuti RA, Agostini E, Iacoangeli M, Rychlicki F, Capecci nidis JP, Krüger R, et al: Collaborative analysis of alpha-synu- M, Gabriella MG, et al: [Deep brain stimulation in Parkin- clein gene promoter variability and Parkinson disease. JAMA son’s disease. Experience in Ancona.] J Neurosurg Sci 47 (1 296:661–670, 2006 Suppl 1):12–16, 2003 (Ital) 54. Maraganore DM, de Andrade M, Lesnick TG, Strain KJ, 73. Romito LM, Contarino MF, Ghezzi D, Franzini A, Garava- Farrer MJ, Rocca WA, et al: High-resolution whole-genome glia B, Albanese A: High frequency stimulation of the sub- association study of Parkinson disease. Am J Hum Genet thalamic nucleus is efficacious in Parkin disease. J Neurol 77:685–693, 2005 252:208–211, 2005 55. Maraganore DM, Lesnick TG, Elbaz A, Chartier-Harlin MC, 74. Saint-Cyr JA, Trépanier LL, Kumar R, Lozano AM, Lang Gasser T, Krüger R, et al: UCHL1 is a Parkinson’s disease AE: Neuropsychological consequences of chronic bilateral susceptibility gene. Ann Neurol 55:512–521, 2004 stimulation of the subthalamic nucleus in Parkinson’s disease. 56. Massey AC, Kaushik S, Sovak G, Kiffin R, Cuervo AM: Con- Brain 123 (Pt 10):2091–2108, 2000 sequences of the selective blockage of chaperone-mediated 75. Schüpbach M, Lohmann E, Anheim M, Lesage S, Czernecki autophagy. Proc Natl Acad Sci U S A 103:5805–5810, 2006 V, Yaici S, et al: Subthalamic nucleus stimulation is effica- 57. Mata IF, Samii A, Schneer SH, Roberts JW, Griffith A, Leis cious in patients with Parkinsonism and LRRK2 mutations. BC, et al: Glucocerebrosidase gene mutations: a risk factor for Mov Disord 22:119–122, 2007 Lewy body disorders. Arch Neurol 65:379–382, 2008 76. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, 58. Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA: Kachergus J, et al: α-Synuclein locus triplication causes Par- LRRK2 in Parkinson’s disease: protein domains and func- kinson’s disease. Science 302:841, 2003 tional insights. Trends Neurosci 29:286–293, 2006 77. Spira PJ, Sharpe DM, Halliday G, Cavanagh J, Nicholson GA: 59. Mitsui J, Mizuta I, Toyoda A, Ashida R, Takahashi Y, Goto J, Clinical and pathological features of a Parkinsonian syndrome et al: Mutations for Gaucher disease confer high susceptibility in a family with an Ala53Thr alpha-synuclein mutation. Ann to Parkinson disease. Arch Neurol 66:571–576, 2009 Neurol 49:313–319, 2001 60. Mizuta I, Satake W, Nakabayashi Y, Ito C, Suzuki S, Momose 78. Tan EK, Khajavi M, Thornby JI, Nagamitsu S, Jankovic J, Y, et al: Multiple candidate gene analysis identifies alpha-sy- Ashizawa T: Variability and validity of polymorphism associ- nuclein as a susceptibility gene for sporadic Parkinson’s dis- ation studies in Parkinson’s disease. Neurology 55:533–538, ease. Hum Mol Genet 15:1151–1158, 2006 2000 61. Nosaka C, Kunimoto S, Atsumi S, Takeuchi T: Inhibition 79. Tan EK, Puong KY, Fook-Chong S, Chua E, Shen H, Yuen Y,

6 Neurosurg Focus / Volume 28 / January 2010

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC Genetic basis of Parkinson disease

et al: Case-control study of UCHL1 S18Y variant in Parkin- molecular causes of Parkinson’s disease. Trends Mol Med son’s disease. Mov Disord 21:1765–1768, 2006 12:521–528, 2006 80. Tan EK, Zhao Y, Skipper L, Tan MG, Di Fonzo A, Sun L, 90. Xiromerisiou G, Hadjigeorgiou GM, Gourbali V, Johnson et al: The LRRK2 Gly2385Arg variant is associated with J, Papakonstantinou I, Papadimitriou A, et al: Screening for Parkinson’s disease: genetic and functional evidence. Hum SNCA and LRRK2 mutations in Greek sporadic and auto- Genet 120:857–863, 2007 somal dominant Parkinson’s disease: identification of two 81. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Har- novel LRRK2 variants. Eur J Neurol 14:7–11, 2007 vey K, Gispert S, et al: Hereditary early-onset Parkinson’s dis- 91. Yang Y, Gehrke S, Imai Y, Huang Z, Ouyang Y, Wang JW, ease caused by mutations in PINK1. Science 304:1158–1160, et al: Mitochondrial pathology and muscle and dopaminer- 2004 gic neuron degeneration caused by inactivation of Drosophi- 82. Valente EM, Salvi S, Ialongo T, Marongiu R, Elia AE, Caputo la Pink1 is rescued by Parkin. Proc Natl Acad Sci U S A V, et al: PINK1 mutations are associated with sporadic early- 103:10793–10798, 2006 onset parkinsonism. Ann Neurol 56:336–341, 2004 92. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, 83. van de Warrenburg BP, Lammens M, Lücking CB, Denèfle P, Ampuero I, et al: The new mutation, E46K, of alpha-synucle- Wesseling P, Booij J, et al: Clinical and pathologic abnormali- in causes Parkinson and Lewy body dementia. Ann Neurol ties in a family with parkinsonism and parkin gene mutations. 55:164–173, 2004 Neurology 56:555–557, 2001 93. Zhang L, Shimoji M, Thomas B, Moore DJ, Yu SW, Marupudi 84. Volles MJ, Lansbury PT Jr: Zeroing in on the pathogenic form NI, et al: Mitochondrial localization of the Parkinson’s dis- of alpha-synuclein and its mechanism of neurotoxicity in Par- ease related protein DJ-1: implications for pathogenesis. Hum kinson’s disease. Biochemistry 42:7871–7878, 2003 Mol Genet 14:2063–2073, 2005 85. West AB, Moore DJ, Choi C, Andrabi SA, Li X, Dikeman D, 94. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln et al: Parkinson’s disease-associated mutations in LRRK2 S, et al: Mutations in LRRK2 cause autosomal-dominant par- link enhanced GTP-binding and kinase activities to neuronal kinsonism with pleomorphic pathology. Neuron 44:601–607, toxicity. Hum Mol Genet 16:223–232, 2007 2004 86. Winkler S, Hagenah J, Lincoln S, Heckman M, Haugarvoll K, 95. Zorzon M, Capus L, Pellegrino A, Cazzato G, Zivadinov R: Lohmann-Hedrich K, et al: α-Synuclein and Parkinson dis- Familial and environmental risk factors in Parkinson’s dis- ease susceptibility. Neurology 69:1745–1750, 2007 ease: a case-control study in north-east Italy. Acta Neurol 87. Wintermeyer P, Krüger R, Kuhn W, Müller T, Woitalla D, Scand 105:77–82, 2002 Berg D, et al: Mutation analysis and association studies of the UCHL1 gene in German Parkinson’s disease patients. Neu­ roreport 11:2079–2082, 2000 Manuscript submitted September 21, 2009. 88. Wong K, Sidransky E, Verma A, Mixon T, Sandberg GD, Accepted October 22, 2009. Wakefield LK, et al: Neuropathology provides clues to the Address correspondence to: Georgios Hadjigeorgiou, M.D., pathophysiology of Gaucher disease. Mol Genet Metab Laboratory of Neurogenetics, Department of Neurology, School of 82:192–207, 2004 Medicine, University of Thessaly, 22 Papakyriazi Street, GR-41222 89. Wood-Kaczmar A, Gandhi S, Wood NW: Understanding the Larissa, Greece. email: [email protected].

Neurosurg Focus / Volume 28 / January 2010 7

Unauthenticated | Downloaded 10/02/21 01:54 AM UTC