5
PERSPECTIVE S58 JULY 2004 NEURODEGENERATION Recent years have seen an explosion in the rate of discovery of genetic defects linked to Parkinson’s disease. These breakthroughs have not provided a direct explanation for the disease process. Nevertheless, they have helped transform Parkinson’s disease research by providing tangible clues to the neurobiology of the disorder. Parkinson’s disease (PD) is the second most common human neu- rodegenerative disorder, after Alzheimer’s dementia. This disease is progressive, with a mean age at onset of 55, and its incidence increases markedly with age 1 . The primary hallmark of PD is the degeneration of the nigrostriatal dopaminergic pathway, which, in depleting the brain of dopamine, initiates aberrant motor activity such as tremor at rest, rigidity, slowness of voluntary movement, and postural instabil- ity. As with other neurodegenerative disorders, however, the neu- ropathology of PD is not restricted to one pathway, and histological abnormalities also occur in many other dopaminergic and non- dopaminergic cell groups including the locus coeruleus, raphe nuclei and nucleus basalis of Meynert 2 . Because numerous distinct neuro- logical conditions share the clinical features of PD, a definitive diag- nosis of PD can only be made at autopsy, and it has customarily been based not only on the loss of nigrostriatal dopaminergic neurons but also on the presence of intraneuronal inclusions called Lewy bodies (LBs). These are spherical eosinophilic cytoplasmic aggregates con- taining a variety of proteins, of which α-synuclein is a major compo- nent 3 , and are found in every affected brain region. Whether identification of LBs should still be considered necessary for the diag- nosis of PD is controversial, in that individuals with inherited PD linked to mutations in the gene encoding parkin typically lack LBs and are still regarded as having PD 1 . Moreover, the role of LBs in the PD neurodegenerative process is a matter of fierce debate. The cause of almost all cases of PD remains unknown. PD generally arises as a sporadic condition but is occasionally inherited as a simple mendelian trait (Table 1). Although sporadic and familial PD are very similar, inherited forms of the disease usually begin at earlier ages and are associated with atypical clinical features (Table 1). Until recently, all of the hypotheses regarding the cause and mechanism of PD neu- rodegeneration derived from investigations carried out on autopsy tissues from individuals with sporadic PD or in neurotoxic animal models such as that produced by the mitochondrial poison 1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) 1 . In the mid-1990s, however, this situation changed with the identification of a mutation in the α-synuclein gene associated with PD in an Italian kindred 4 . Since then, four additional genetic defects underlying PD have been identified and linkages have been reported for at least four more (Table 1). Here we review what is currently known about these PD-causing mutations. As is the case in Alzheimer’s disease, these gene defects seem to operate on a common molecular pathway. Thus, we also dis- cuss this pathway and the directions in which those genes may lead us in regard to the development of genetically based animal models, which are crucial to unraveling the basis of the neurodegenerative processes of PD. α-Synuclein mutations and dopaminergic neurodegeneration Three missense mutations (A53T,A30P and E46K) in the gene encod- ing α-synuclein are linked to a dominantly inherited PD 4–6 (Table 1). None of these mutations has been found in sporadic PD or in individ- uals without the disease. Injection of either human wild-type or mutant α-synuclein- expressing viral vectors into the rat and monkey nigrostriatal pathway causes dopaminergic neurodegeneration associated with α−synu- clein-containing inclusions 7,8 . Transgenic overexpression of mutant or wild-type α-synuclein in mice or flies has produced equivocal results 1 , however, in that intraneuronal proteinaceous inclusions, but not definite neuronal death, have generally been documented. Still, these results, together with the finding that α-synuclein ablation in mice does not cause neurodegeneration 9,10 , support the notion that α-synuclein mutations operate by a toxic gain-of-function mecha- nism. Viral vector–mediated overexpression of wild-type α-synuclein reproduces PD neuropathology in animals 7,8 , and genomic multipli- cation of the gene encoding α-synuclein is associated with a familial form of PD 11,12 . It is thus possible that the function gained by the mutant protein is not a newly acquired property, but rather a native property that is enhanced and becomes deleterious. How mutant α-synuclein variants produce neurotoxicity remains elusive, in part because the protein’s function is just beginning to be understood. Wild-type α-synuclein binds preferentially to plasma membranes (rather than mitochondrial membranes) in yeasts 13 and this interaction, which is mediated by major conformational changes of the protein 14 , seems to be crucial to several of its physiological functions 1 . Membrane-bound α-synuclein has been proposed to modulate phospholipase D activity 15 , thereby perhaps influencing the availability of synaptic vesicles for release. Membrane-bound α-synu- Genetic clues to the pathogenesis of Parkinson’s disease Miquel Vila & Serge Przedborski Miquel Vila is in the Department of Neurology and Serge Przedborski is in the Departments of Neurology and Pathology and the Center for Neurobiology and Behavior, Columbia University, New York, New York 10032, USA. e-mail: [email protected] Published online 1 July 2004; doi: 10.1038/nm1068 © 2004 Nature Publishing Group http://www.nature.com/naturemedicine

Genetic clues to the pathogenesis of Parkinson's disease

  • Upload
    serge

  • View
    218

  • Download
    2

Embed Size (px)

Citation preview

Page 1: Genetic clues to the pathogenesis of Parkinson's disease

P E R S P E C T I V E

S58 JULY 2004 NEURODEGENERATION

Recent years have seen an explosion in the rate of discovery ofgenetic defects linked to Parkinson’s disease. Thesebreakthroughs have not provided a direct explanation for thedisease process. Nevertheless, they have helped transformParkinson’s disease research by providing tangible clues to theneurobiology of the disorder.

Parkinson’s disease (PD) is the second most common human neu-rodegenerative disorder, after Alzheimer’s dementia. This disease isprogressive, with a mean age at onset of 55, and its incidence increasesmarkedly with age1. The primary hallmark of PD is the degenerationof the nigrostriatal dopaminergic pathway, which, in depleting thebrain of dopamine, initiates aberrant motor activity such as tremor atrest, rigidity, slowness of voluntary movement, and postural instabil-ity. As with other neurodegenerative disorders, however, the neu-ropathology of PD is not restricted to one pathway, and histologicalabnormalities also occur in many other dopaminergic and non-dopaminergic cell groups including the locus coeruleus, raphe nucleiand nucleus basalis of Meynert2. Because numerous distinct neuro-logical conditions share the clinical features of PD, a definitive diag-nosis of PD can only be made at autopsy, and it has customarily beenbased not only on the loss of nigrostriatal dopaminergic neurons butalso on the presence of intraneuronal inclusions called Lewy bodies(LBs). These are spherical eosinophilic cytoplasmic aggregates con-taining a variety of proteins, of which α-synuclein is a major compo-nent3, and are found in every affected brain region. Whetheridentification of LBs should still be considered necessary for the diag-nosis of PD is controversial, in that individuals with inherited PDlinked to mutations in the gene encoding parkin typically lack LBsand are still regarded as having PD1. Moreover, the role of LBs in thePD neurodegenerative process is a matter of fierce debate.

The cause of almost all cases of PD remains unknown. PD generallyarises as a sporadic condition but is occasionally inherited as a simplemendelian trait (Table 1). Although sporadic and familial PD are verysimilar, inherited forms of the disease usually begin at earlier ages andare associated with atypical clinical features (Table 1). Until recently,all of the hypotheses regarding the cause and mechanism of PD neu-rodegeneration derived from investigations carried out on autopsytissues from individuals with sporadic PD or in neurotoxic animal

models such as that produced by the mitochondrial poison 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)1. In the mid-1990s,however, this situation changed with the identification of a mutationin the α-synuclein gene associated with PD in an Italian kindred4.Since then, four additional genetic defects underlying PD have beenidentified and linkages have been reported for at least four more(Table 1).

Here we review what is currently known about these PD-causingmutations. As is the case in Alzheimer’s disease, these gene defectsseem to operate on a common molecular pathway. Thus, we also dis-cuss this pathway and the directions in which those genes may lead usin regard to the development of genetically based animal models,which are crucial to unraveling the basis of the neurodegenerativeprocesses of PD.

α-Synuclein mutations and dopaminergic neurodegenerationThree missense mutations (A53T, A30P and E46K) in the gene encod-ing α-synuclein are linked to a dominantly inherited PD4–6 (Table 1).None of these mutations has been found in sporadic PD or in individ-uals without the disease.

Injection of either human wild-type or mutant α-synuclein-expressing viral vectors into the rat and monkey nigrostriatal pathwaycauses dopaminergic neurodegeneration associated with α−synu-clein-containing inclusions7,8. Transgenic overexpression of mutantor wild-type α-synuclein in mice or flies has produced equivocalresults1, however, in that intraneuronal proteinaceous inclusions, butnot definite neuronal death, have generally been documented. Still,these results, together with the finding that α-synuclein ablation inmice does not cause neurodegeneration9,10, support the notion thatα-synuclein mutations operate by a toxic gain-of-function mecha-nism. Viral vector–mediated overexpression of wild-type α-synucleinreproduces PD neuropathology in animals7,8, and genomic multipli-cation of the gene encoding α-synuclein is associated with a familialform of PD11,12. It is thus possible that the function gained by themutant protein is not a newly acquired property, but rather a nativeproperty that is enhanced and becomes deleterious.

How mutant α-synuclein variants produce neurotoxicity remainselusive, in part because the protein’s function is just beginning to beunderstood. Wild-type α-synuclein binds preferentially to plasmamembranes (rather than mitochondrial membranes) in yeasts13 andthis interaction, which is mediated by major conformational changesof the protein14, seems to be crucial to several of its physiologicalfunctions1. Membrane-bound α-synuclein has been proposed tomodulate phospholipase D activity15, thereby perhaps influencing theavailability of synaptic vesicles for release. Membrane-bound α-synu-

Genetic clues to the pathogenesis of Parkinson’s diseaseMiquel Vila & Serge Przedborski

Miquel Vila is in the Department of Neurology and Serge Przedborski is in theDepartments of Neurology and Pathology and the Center for Neurobiology andBehavior, Columbia University, New York, New York 10032, USA.e-mail: [email protected]

Published online 1 July 2004; doi: 10.1038/nm1068

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 2: Genetic clues to the pathogenesis of Parkinson's disease

P E R S P E C T I V E

NEURODEGENERATION JULY 2004 S59

clein also seems to be in dynamic equilibrium with cytosolic α-synu-clein13, which upon accumulation can render endogenous dopaminetoxic16, and to act as a seed promoting the formation of cytosolicinclusions17. Conceivably if these aggregates are not promptly clearedby degradation pathways18, neurotoxicity can ensue.

Both wild-type and mutant α-synuclein form amyloid fibrils akinto those seen in LBs, as well as nonfibrillary oligomers1 termedprotofibrils. Because the pathogenic α-synucleinA53T mutant pro-motes the formation of protofibrils19, these oligomers may be thetoxic species of α-synuclein. In keeping with this and with the knownassociation of α-synuclein with synaptosomes, protofibrils may causetoxicity by permeabilizing synaptic vesicles20, allowing dopamine toleak into the cytoplasm and participate in reactions that generateoxidative stress. Furthermore, the selective vulnerability of nigrostri-atal neurons in PD may derive from the ability of dopamine ordopamine-quinone to stabilize α-synuclein protofibrils21. However,protofibrils have only been identified and studied in vitro, and so fur-ther work is required to establish whether they form in neurons andwhether their formation correlates with neurotoxicity.

Parkin, a protein with many substratesLoss-of-function mutations in the gene encoding parkin cause arecessively inherited form of PD22 (Table 1). The onset of parkin-related PD usually, but not always, occurs before age 30 (ref. 1).Pathologically, this form of familial PD is associated with a loss ofnigrostriatal neurons, but LBs are not typically observed. Parkin-nullmice and flies do not develop degeneration of nigrostriatal dopamin-ergic neurons23–25. However, these animals do show functional mito-chondrial deficits24–26 suggestive of those seen in sporadic PD1.

Identification of the normal function of parkin has provided hintsto the pathogenic effects of parkin mutations. Parkin is one of a classof proteins containing two RING-FINGER DOMAINS separated by an in-between RING-finger domain, and like other such proteins, parkinfunctions as an E3 ubiquitin ligase27,28, a component of the ubiquitin

system. Many mutations affecting parkin abolish its E3 ligase activ-ity1, as does the post-translational modification (S-nitrosylation) ofwild-type parkin29. It is thus conceivable that parkin dysfunction isinvolved in the pathogenesis of both familial and sporadic PD, but theunderlying molecular details remain speculative. Loss of parkin activ-ity may trigger cell death by rendering neurons more susceptible tocytotoxic insults, such as those caused by proteasome inhibition ormutant α-synuclein30, or by impairing ubiquitination of cyclin E31, amolecule previously implicated in neuronal apoptosis. In support ofthe latter hypothesis, cyclin E is abundant in the midbrains of individ-uals with parkin-related PD, and overexpression of wild-type parkinattenuates cyclin E accumulation and promotes survival in excito-toxin-treated cultured neurons31. Several studies have shown func-tional interactions between parkin and α-synuclein, and havesuggested that these interactions may involve the proteasome1. Otherinvestigations have highlighted the multiplicity of parkin substratesand how these might have a key role in neuronal death1. However,none of the parkin substrates that have been identified seem to bespecifically enriched in dopaminergic neurons. Thus, further studiesare needed to explain the relative specificity of dopaminergic neu-rodegeneration mediated by parkin mutations.

UCH-L1 dabbles in degenerationUbiquitin C-terminal hydrolase-L1 (UCH-L1) is expressed mainlyin the brain, where it catalyzes the hydrolysis of C-terminal ubiqui-tyl esters. A single dominant mutant form (I93M) of UCH-L1,found in two members of a PD-affected family, has been implicatedin the development of an inherited form of PD32. Conversely, it hasbeen confirmed that a polymorphism (S18Y) of UCH-L1 reducesthe risk of developing sporadic PD, especially in early-onset cases33.The I93M mutation decreases the enzyme’s activity, suggesting thata loss of function is the culprit in disease development. However,mice carrying a UCH-L1-null mutation do show neurodegenerativechanges, but not in the nigrostriatal dopaminergic pathway34. Upon

Table 1 Genes and loci linked to familial PD

Locus Chromosomal location Protein Inheritance pattern Atypical PD features Lewy bodies

PARK1 4q21 α-Synucleina AD Early onset YesLower prevalence of tremor

PARK2 6q25.2–q27 Parkin AR Early or juvenile onset Mostly negativeb

More frequent dystonia and levodopa-induced dyskinesiasSlower disease progression

PARK3 2p13 Unknown AD Dementia in some individuals YesRapid progression

PARK4c 4p15 Unknown AD Early onset YesRapid progressionDementiaAutonomic dysfunctionPostural tremor

PARK5 4p14 UCH-L1 AD None Unknown

PARK6 1p36 PINK1 AR Early onset UnknownSlow progression

PARK7 1p36 DJ-1 AR Early onset UnknownPsychiatric symptomsSlow progression

PARK8 12p11.2–q13.1 Unknown AD None No

PARK9 1p36 Unknown AR Juvenile onset UnknownSpasticitySupranuclear gaze paralysisDementia

AD, autosomal dominant; AR, autosomal recessive. aIncluding mutations and wild-type multiplications. bLewy bodies reported in one individual with parkin mutations54. cTheinitial PARK4 linkage to 4p15 could not be confirmed, and the PD phenotype in this family was subsequently linked to a PARK1 variant (α-synuclein triplication)11.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 3: Genetic clues to the pathogenesis of Parkinson's disease

P E R S P E C T I V E

S60 JULY 2004 NEURODEGENERATION

dimerization, UCH-L1 can also exert an ubiquitin ligase activity,which is decreased by the pathogenic I93M mutation and increasedby the protective S18Y polymorphism35. UCH-L1 was also identi-fied as a protease with specificity for Nedd8 (ref. 36), a small ubiqui-tin-like protein implicated in cell-cycle regulation. Although all ofthese findings are consistent with the hypothesis that PD pathogen-esis involves impairment of the ubiquitin systems, further studiesare needed to elucidate how mutations affecting UCH-L1 killdopaminergic neurons.

Multiplicity of DJ-1 mutationsDJ-1 is a homodimeric, multifunctional protein ubiquitouslyexpressed in human tissues including the brain. Eleven differentmutations affecting DJ-1—these include missense, truncating andsplice-site mutations and large deletions—have been linked to anautosomal recessive form of PD37–39. Although DJ-1 mutationsaccount for only a small fraction of early-onset PD, they are the sec-ond most frequent cause of recessive forms of PD after parkin muta-tions. These mutations are found throughout the DJ-1 gene in four ofits seven exons. This suggests that even though linked to a similar phe-notype, the different mutations probably affect different domains ofthe protein with distinct functional andstructural roles. This situation is reminiscentof that in amyotrophic lateral sclerosis, wherethe same paralytic syndrome is caused bymany different mutations affecting super-oxide dismutase-1 (http://alsod.org).

DJ-1 was discovered as part of a multipro-tein complex that stabilizes mRNA through aninteraction with c-myc and as a proteininvolved in infertility in rodents40. DJ-1 maymodulate mRNA expression through interac-tions with a polypeptide complex comprisingthe androgen receptor and the SUMOYLATION

enzyme PIASx41,42. DJ-1 may also functionboth as a sensor for oxidative stress and as anantioxidant40,43. In addition, structural studiesindicate that DJ-1 shares similarities with bac-terial Hsp31, a stress-inducible chaperone44.Yet none of this information sheds much lightinto the pathogenic mechanisms of DJ-1mutations in PD. Studies of a single DJ-1mutant, L166P, have shown that it hasimpaired folding properties and cannot form ahomodimer45,46. Thus, as a misfolded and lessstable protein, DJ-1L166P may cause cytotoxic-ity by overwhelming the cellular protein degra-dation systems and by undergoing abnormalsubcellular localization, for instance in mito-chondria45. It remains to be established howaccurate this pathogenic scenario is andwhether DJ-1L166P reliably reflects the molecu-lar abnormalities of the other known DJ-1mutations. Furthermore, DJ-1 chiefly localizesin brain glial cells47, suggesting that neuronaldeath mediated by DJ-1 mutations may arisefrom a NON-CELL-AUTONOMOUS process.

PINK1, the new kid on the blockAUTOZYGOSITY mapping of a large consan-guineous Sicilian family localized the PARK6

locus, which is linked to an autosomal recessive form of PD, to chromo-some 1p35–36 (ref. 48). PARK6 has since been linked to an early-onsetrecessive PD in eight additional families from four European coun-tries49. Sequencing of candidate genes within the PARK6 region inaffected members from each family identified two homozygous muta-tions in the gene encoding PTEN-induced putative kinase-1 (PINK1,also known as BRPK)50. PINK1 is a 581-amino-acid protein with a pre-dicted molecular mass of 62.8 kDa, which shows 95% sequence similar-ity between human and mouse51,52. In adult mice, PINK1 isubiquitously expressed among tissues, with an apparently high expres-sion in the brain51. In both human and mouse PINK1, a serine/threo-nine kinase domain is the sole known functional domain51,52.Presumably PINK1 also has a mitochondrial targeting motif, consistentwith the observation that it localizes to the mitochondria in transfectedcell lines50. If confirmed, this finding may be significant, as a defect inmitochondrial function has been implicated in the pathogenesis of PD1.

Of the two identified mutations, one is a missense mutation(G309D) in the putative kinase domain and the second is a nonsensemutation (W437OPA) truncating the last 145 amino acids of thekinase domain C terminus50. Both mutations are expected to impairPINK1 kinase activity or substrate recognition and to cause PD

Normal proteinMisfolded protein

DJ-1mutation

α-Synucleinmutation

Parkinmutation

UCH-L1mutation

Proteasome

Lysosome

Protein degradationpathways

Cell death

Degradedproteins

Modifiedparkin

Modifiedα-synuclein DJ-1/PINK1

mutations

DJ-1mutation

Mitochondrialdysfunction

Dopamine

ROS

Detoxification

Figure 1 Genetic mutations and the pathogenesis of PD. Misfolded proteins may contribute to PDneurodegeneration. Mutant α-synuclein and DJ-1 may be misfolded (blue arrows), thus overloadingthe ubiquitin (proteasomal) and lysosomal degradation pathways. Other mutant proteins, such asparkin and UCH-L1, may lack their wild-type function. Both of these proteins, which belong to theubiquitin-proteasome system, upon mutation may no longer exert their ubiquitin ligase activity, thusdamaging the ability of the cellular machinery to detect and degrade misfolded proteins (red arrows).Mutations in DJ-1 may also alter its supposed chaperone activity, disrupting the refolding of damagedproteins or the targeting and delivery of damaged proteins for degradation (red arrows). Thesedifferent alterations may lead to the accumulation of unwanted proteins, which, by unknownmechanisms (dashed arrows), may lead to neurodegeneration. Oxidative stress generated bymitochondrial dysfunction and dopamine metabolism may also promote protein misfolding as a resultof post-translational modifications, especially of α-synuclein and parkin. Oxidative stress in PD mayalso originate from a defect in the reduced capacity of DJ-1 to detoxify reactive oxygen species,whereas the mitochondrial dysfunction may, at least in part, derive from defective activity andmislocation of DJ-1 and PINK1. Mitochondrial dysfunction, oxidative stress and protein mishandlingare thus tightly interconnected in this hypothesized pathogenic cascade. Additional possibleinteractions have been omitted for clarity.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 4: Genetic clues to the pathogenesis of Parkinson's disease

P E R S P E C T I V E

NEURODEGENERATION JULY 2004 S61

through a loss of function. Yet reduced striatal [18F]dopa uptakerevealed by positron emission tomography has been found in asymp-tomatic PARK6 heterozygotes, raising the possibility that PINK1mutations may operate through haploinsufficiency or a dominant-negative effect53. In either case, impaired phosphorylation of PINK1’ssubstrate, especially in mitochondria, is a likely scenario for the pathogenic mechanism of the two mutations.

Although the crucial substrates of PINK1 have yet to be identified,we already know that neuroblastoma cells transiently transfected witheither wild-type or mutant PINK1 do not show detectable alterationsin viability50. In contrast, when these cells are challenged with theproteasome inhibitor MG132, overexpression of wild-type PINK1mitigates cell death, whereas overexpression of mutant PINK1 neitherattenuates nor enhances MG132-mediated cytotoxicity50. Theseresults suggest that the loss of PINK1 function renders dopaminergicneurons more vulnerable to injury. This possibility does fit neatly intothe concept that interactions between genetic and environmental fac-tors may be responsible for the neurodegeneration in sporadic PD1.

ConclusionThe shared phenotype associated with the different genetic mutationswe have discussed raises the tantalizing possibility of a molecularintersection in the pathogenic mechanisms driven by these distinctPD-causing mutations (Fig. 1). Among various plausible mechanistichypotheses, available data favor impaired protein degradation andaccumulation of misfolded proteins as the unifying factor linkinggenetic alterations to dopaminergic neurodegeneration in familial PD(Fig. 1). According to this reasoning, α-synuclein and DJ-1 mutationswould cause abnormal protein conformations, overwhelming themain cellular protein degradation systems—the proteasomal andlysosomal pathways—whereas parkin and UCH-L1 mutations wouldundermine the cell’s ability to detect and degrade misfolded proteins.The common end result of these different perturbations is thusexpected to be a cellular buildup of unwanted proteins that shouldhave been cleared. Minimal defects in this protein turnover machin-ery may suffice to cause a slow demise of dopaminergic neurons,which may explain the relentless, progressive nature of the disease.This scenario does not, however, explain why an accumulation ofmisfolded proteins, which is likely to occur in all cells, would inflict

greater damage on dopaminergic neurons in familial PD. Perhapsnigrostriatal dopaminergic neurons are less able to cope with ‘mis-folded protein stress’ because of a higher basal load of damaged pro-teins that is due to dopamine-mediated oxidative events. Also poorlyaddressed by the above scenario is the link between previously identi-fied factors in PD neurodegeneration, such as mitochondrial dysfunc-tion or oxidative stress, and the molecular events engendered by thePD-causing mutations. The hypothesized mitochondrial location ofDJ-1 and PINK1 and the role of DJ-1 in oxidative stress may emergeas crucial in efforts to reconcile the different aspects of the unifiedpathogenic cascade of PD.

ACKNOWLEDGMENTSThe authors thank R.E. Burke, D. Sulzer and W. Dauer for insightful comments onthe manuscript, M.J. Farrer and A.B. Singleton for input concerning PARK4, andM. Lucas for assistance in preparing this manuscript. The authors are supported byNIH/NINDS (grants R29 NS37345, RO1 NS38586 and NS42269, P50 NS38370,and P01 NS11766-27A1), NIH/NIA grant (RO1 AG021617-01), the USDepartment of Defense (DAMD 17-99-1-9471, DAMD 17-03-1 and DAMD17-03-1-0428), the Lowenstein Foundation, the Lillian Goldman Charitable Trust, theParkinson’s Disease Foundation, the American Parkinson Disease Association, andMDA/Wings-Over-Wall Street.

COMPETING INTERESTS STATEMENTThe authors declare that they have no competing financial interest.

HOW TO CITE THIS ARTICLEPlease cite this article as supplement to volume 10 of Nature Medicine, pagesS58–S62.

Published online at http://www.nature.com/focus/neurodegen/

1. Dauer, W. & Przedborski, S. Parkinson’s disease: mechanisms and models. Neuron39, 889–909 (2003).

2. Braak, H. et al. Staging of brain pathology related to sporadic Parkinson’s disease.Neurobiol. Aging 24, 197–211 (2003).

3. Spillantini, M.G. et al. α-Synuclein in Lewy bodies. Nature 388, 839–840 (1997).4. Polymeropoulos, M.H. et al. Mutation in the α-synuclein gene identified in families

with Parkinson’s disease. Science 276, 2045–2047 (1997).5. Kruger, R. et al. Ala30Pro mutation in the gene encoding α-synuclein in Parkinson’s

disease. Nat. Genet. 18, 107–108 (1998).6. Zarranz, J.J. et al. The new mutation, E46K, of α-synuclein causes Parkinson and

Lewy body dementia. Ann. Neurol. 55, 164–173 (2004).7. Lo Bianco, C., Ridet, J.L., Schneider, B.L., Deglon, N. & Aebischer, P. α-

Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-basedmodel of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 99, 10813–8 (2002).

8. Kirik, D. et al. Nigrostriatal α-synucleinopathy induced by viral vector-mediatedoverexpression of human α-synuclein: a new primate model of Parkinson’s disease.Proc. Natl. Acad. Sci. USA 100, 2884–2889 (2003).

9. Abeliovich, A. et al. Mice lacking α-synuclein display functional deficits in thenigrostriatal dopamine system. Neuron 25, 239–252 (2000).

10. Dauer, W. et al. Resistance of α-synuclein null mice to the parkinsonian neurotoxinMPTP. Proc. Natl. Acad. Sci. USA 99, 14524–14529 (2002).

11. Singleton, A.B. et al. α-Synuclein locus triplication causes Parkinson’s disease.Science 302, 841 (2003).

12. Chartier-Harlin, M.C. et al. α-Synuclein locus duplication causes familialParkinson’s disease. Lancet (in the press).

13. Outeiro, T.F. & Lindquist, S. Yeast cells provide insight into α-synuclein biology andpathobiology. Science 302, 1772–1775 (2003).

14. Jao, C.C., Der-Sarkissian, A., Chen, J. & Langen, R. Structure of membrane-bound α-synuclein studied by site-directed spin labeling. Proc. Natl. Acad. Sci. USA 101,8331–8336 (2004).

15. Payton, J.E., Perrin, R.J., Woods, W.S. & George, J.M. Structural determinants ofPLD2 inhibition by α-synuclein. J. Mol. Biol. 337, 1001–1009 (2004).

16. Xu, J.et al. Dopamine-dependent neurotoxicity of α-synuclein: a mechanism forselective neurodegeneration in Parkinson disease. Nat. Med. 8, 600–606 (2002).

17. Lee, H.J., Choi, C. & Lee, S.J. Membrane-bound α-synuclein has a high aggregationpropensity and the ability to seed the aggregation of the cytosolic form. J. Biol. Chem.277, 671–678 (2002).

18. Lee, H. J., Khoshaghideh, F., Patel, S. & Lee, S. J. Clearance of α-synucleinoligomeric intermediates via the lysosomal degradation pathway. J. Neurosci. 24,1888–1896 (2004).

19. Conway, K. A. et al. Acceleration of oligomerization, not fibrillization, is a sharedproperty of both α-synuclein mutations linked to early-onset Parkinson’s disease:implications for pathogenesis and therapy. Proc. Natl. Acad. Sci. USA 97, 571–576(2000).

GLOSSARYRING domain One of a class of protein domains that consist oftwo loops that are held together at their base by cysteine andhistidine residues that complex two zinc ions. Proteins containingdomains of this type are known as RING-finger proteins.

Sumoylation The attachment of SUMO, a ubiquitin-like modifierprotein. But in contrast to ubiquitin, which targets proteins fordegradation, SUMO seems to affect the subcellular localization ofproteins and enhance their stability.

Non-cell-autonomous A genetic trait in which genotypicallymutant cells cause other cells (regardless of their genotype) toshow a mutant phenotype. By contrast, a cell-autonomous trait isone in which only genotypically mutant cells show the mutantphenotype.

Autozygosity Homozygosity by virtue of parental descent from acommon ancestor.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 5: Genetic clues to the pathogenesis of Parkinson's disease

P E R S P E C T I V E

S62 JULY 2004 NEURODEGENERATION

20. Volles, M.J. et al. Vesicle permeabilization by protofibrillar α-synuclein: implicationsfor the pathogenesis and treatment of Parkinson’s disease. Biochemistry 40,7812–7819 (2001).

21. Conway, K.A., Rochet, J.C., Bieganski, R.M. & Lansbury, P.T. Jr. Kinetic stabilizationof the α-synuclein protofibril by a dopamine–α-synuclein adduct. Science 294,1346–1349 (2001).

22. Kitada, T. et al. Mutations in the parkin gene cause autosomal recessive juvenileparkinsonism. Nature 392, 605–608 (1998).

23. Goldberg, M.S. et al. Parkin-deficient mice exhibit nigrostriatal deficits but not lossof dopaminergic neurons. J. Biol. Chem. 278, 43628–43635 (2003).

24. Pesah, Y. et al. Drosophila parkin mutants have decreased mass and cell size andincreased sensitivity to oxygen radical stress. Development 131, 2183–2194 (2004).

25. Greene, J. C. et al. Mitochondrial pathology and apoptotic muscle degeneration inDrosophila parkin mutants. Proc. Natl. Acad. Sci. USA 100, 4078–4083 (2003).

26. Palacino, J.J. et al. Mitochondrial dysfunction and oxidative damage in parkin-defi-cient mice. J. Biol. Chem. 279, 18614–18622 (2004).

27. Shimura, H. et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-pro-tein ligase. Nat. Genet. 25, 302–305 (2000).

28. Zhang, Y. et al. Parkin functions as an E2-dependent ubiquitin- protein ligase and pro-motes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc. Natl.Acad. Sci. USA 97, 13354–13359 (2000).

29. Chung, K.K. et al. S-Nitrosylation of parkin regulates ubiquitination and compromisesparkin’s protective function. Science 304, 1328–1331 (2004).

30. Petrucelli, L. et al. Parkin protects against the toxicity associated with mutant α-synu-clein: proteasome dysfunction selectively affects catecholaminergic neurons. Neuron36, 1007–1019 (2002).

31. Staropoli, J.F. et al. Parkin is a component of an SCF-like ubiquitin ligase complex andprotects postmitotic neurons from kainate excitotoxicity. Neuron 37, 735–749(2003).

32. Leroy, E. et al. The ubiquitin pathway in Parkinson’s disease Nature 55, 512–521(2004).

33. Maraganore, D.M., et al. UCHL1 is a Parkinson's disease susceptibility gene. Ann.Neurol. 55, 512–521 (2004).

34. Saigoh, K. et al. Intragenic deletion in the gene encoding ubiquitin carboxy-terminalhydrolase in gad mice. Nat. Genet. 23, 47–51 (1999).

35. Liu, Y., Fallon, L., Lashuel, H.A., Liu, Z. & Lansbury, P.T. Jr. The UCH-L1 gene encodestwo opposing enzymatic activities that affect α-synuclein degradation and Parkinson’sdisease susceptibility. Cell 111, 209–218 (2002).

36. Hemelaar, J. et al. Specific and covalent targeting of conjugating and deconjugatingenzymes of ubiquitin-like proteins. Mol. Cell Biol. 24, 84–95 (2004).

37. Bonifati, V. et al. Mutations in the DJ-1 gene associated with autosomal recessive

early-onset parkinsonism. Science 299, 256–259 (2003).38. Hague, S. et al. Early-onset Parkinson’s disease caused by a compound heterozygous

DJ-1 mutation. Ann. Neurol. 54, 271–274 (2003).39. Abou-Sleiman, P.M., Healy, D.G., Quinn, N., Lees, A.J. & Wood, N.W. The role of path-

ogenic DJ-1 mutations in Parkinson’s disease. Ann. Neurol. 54, 283–286 (2003).40. Cookson, M.R. Pathways to Parkinsonism. Neuron 37, 7–10 (2003).41. Niki, T., Takahashi-Niki, K., Taira, T., Iguchi-Ariga, S.M. & Ariga, H. DJBP: a novel

DJ-1-binding protein, negatively regulates the androgen receptor by recruiting his-tone deacetylase complex, and DJ-1 antagonizes this inhibition by abrogation ofthis complex. Mol. Cancer Res. 1, 247–261 (2003).

42. Takahashi, K. et al. DJ-1 positively regulates the androgen receptor by impairingthe binding of PIASx α to the receptor. J. Biol. Chem. 276, 37556–37563 (2001).

43. Taira, T. et al. DJ-1 has a role in antioxidative stress to prevent cell death. EMBORep. 5, 213–218 (2004).

44. Quigley, P.M., Korotkov, K., Baneyx, F. & Hol, W.G. The 1.6-A crystal structure ofthe class of chaperones represented by Escherichia coli Hsp31 reveals a putativecatalytic triad. Proc. Natl. Acad. Sci. USA 100, 3137–3142 (2003).

45. Moore, D.J., Zhang, L., Dawson, T.M. & Dawson, V.L. A missense mutation (L166P)in DJ-1, linked to familial Parkinson’s disease, confers reduced protein stabilityand impairs homo-oligomerization. J. Neurochem. 87, 1558–1567 (2003).

46. Olzmann, J.A. et al. Familial Parkinson’s disease-associated L166P mutation dis-rupts DJ-1 protein folding and function. J. Biol. Chem. 279, 8506–8515 (2004).

47. Bandopadhyay, R. et al. The expression of DJ-1 (PARK7) in normal human CNSand idiopathic Parkinson’s disease. Brain 127, 420–430 (2004).

48. Valente, E.M. et al. Localization of a novel locus for autosomal recessive early-onset parkinsonism, PARK6, on human chromosome 1p35-p36. Am. J. Hum.Genet. 68, 895–900 (2001).

49. Valente, E.M. et al. PARK6-linked parkinsonism occurs in several European fami-lies. Ann. Neurol. 51, 14–18 (2002).

50. Valente, E.M. et al. Hereditary early-onset parkinson’s disease caused by muta-tions in PINK1. Science 304, 1158–1160 (2004).

51. Nakajima, A., Kataoka, K., Hong, M., Sakaguchi, M. & Huh, N.H. BRPK, a novelprotein kinase showing increased expression in mouse cancer cell lines withhigher metastatic potential. Cancer Lett. 201, 195–201 (2003).

52. Unoki, M. & Nakamura, Y. Growth-suppressive effects of BPOZ and EGR2, twogenes involved in the PTEN signaling pathway. Oncogene 20, 4457–4465(2001).

53. Khan, N.L. et al. Clinical and subclinical dopaminergic dysfunction in PARK6-linked parkinsonism: an 18F-dopa PET study. Ann. Neurol. 52, 849–853 (2002).

54. Farrer, M. et al. Lewy bodies and parkinsonism in families with parkin mutations.Ann. Neurol. 50, 293–300 (2001).

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine