Articles

  • the Korean Society for Brain and Neural Sciences

Article

Review Article

Exp Neurobiol 2013; 22(4): 249-257

Published online December 30, 2013

https://doi.org/10.5607/en.2013.22.4.249

© The Korean Society for Brain and Neural Sciences

LRRK2 as a Potential Genetic Modifier of Synucleinopathies: Interlacing the Two Major Genetic Factors of Parkinson’s Disease

Cheol Hwan Hyun2#, Chae Young Yoon2#, He-Jin Lee2,3 and Seung-Jae Lee1,2*

1Department of Biomedical Science and Technology, 2Institute of Biomedical Science and Technology, 3Department of Anatomy, School of Medicine, Konkuk University, Seoul 143-701, Korea

Correspondence to: *To whom correspondence should be addressed.
TEL: 82-2-450-4166, FAX: 82-2-447-5683
e-mail: sjlee@konkuk.ac.kr
#These authors contributed equally to this work (alphabetical order).

Received: December 1, 2013; Revised: December 13, 2013; Accepted: December 13, 2013

Parkinson's disease (PD) and related Lewy body diseases are characterized by deposition of α-synuclein aggregates in both the central nervous system and peripheral nervous system. Synucleinopathy lesions spread to larger brain areas as the disease progresses, and prion-like cell-to-cell transmission of aggregated α-synuclein is thought to be the underlying mechanism for this pathological spreading. LRRK2 is another protein linked to the pathogenesis of PD, and its presence in Lewy bodies has attracted much attention as to whether LRRK2 and α-synuclein interplay during the pathogenesis of PD. However, the relationship between these two crucial proteins still remains unclear. In this review article, we will discuss the current state of knowledge in terms of how these proteins cause the disease and provide the hypothetical mechanisms by which LRRK2 might modify the generation and progression of synucleinopathy.

Keywords: Parkinson’s disease, LRRK2, alpha-synuclein, synucleinopathy, transmission, neurodegeneration

Parkinson's disease (PD) is one of the most common neurode-generative diseases, with clinical symptoms of resting tremor, increased muscle tone, bradykinesia, and abnormal postural righting reflexes [1]. Pathologically, PD is characterized by loss of dopaminergic neurons in the Substantia nigra pars compacta, deposition of α-synuclein visualized in the forms of Lewy bodies and Lewy neurites, and neuroinflammation demonstrated by the activation of microglia [2]. Lewy body inclusions are primarily composed of misfolded/aggregated α-synuclein [3], and these pathological protein aggregates spread to different brain regions in a highly sequential way as the disease progresses [4]. Therefore, identifying factors that contribute to the formation and spreading of α-synuclein aggregates may be critical to the overall understanding of pathogenesis of PD.

α-Synuclein (SNCA) and LRRK2 genes harbor autosomal dominant mutations that cause familial PD with clinical characteristics similar to sporadic PD [5, 6]. Although some have argued that each factor operates independently [7], there might be interactions between the two genes in pathogenesis of PD. Here, we review the recent literature suggesting cooperation between α-synuclein and LRRK2 and propose potential mechanisms underlying this interaction.

In 1996, researchers discovered genetic linkage to chromosome 4 markers in a large group of Greek/Italian descendants [8]. In this kindred and later in several others, SNCA gene (PARK1) displayed a missense mutation A53T [8] and other missense mutations A30P and E46K [9, 10] with largely similar phenotypes but seemingly worse presentation of dementia for E46K [10], as well as duplications [11] and triplications with varying penetrance [6, 12]. Several years after the first report of SNCA mutation, Funayama and colleagues found genetic linkage to chromosome 12 markers in a Japanese family with autosomal dominant parkinsonism [13]. And in 2004, the mutations were located in LRRK2 (PARK8) in chromosome 12 as a cause of PD and also detected in a number of other kindreds [14, 15]. Among the autosomal dominant mutations in LRRK2, G2019S is the most common mutation throughout ethnic groups [16], and is found most frequently in North-African Arabs and Ashkenazi Jews [5, 17].

Autosomal recessive causes of PD have also been linked to several genes: parkin (PARK2) [18], DJ-1 (PARK6) [19, 20], PINK1 (PARK7) [21], and ATP13A2 (PARK9) [22].

Recently, the genome-wide association studies have identified both SNCA and LRRK2 as strong genetic risk loci for sporadic PD, suggesting the roles of these two genes in the pathogenesis of idiopathic PD [6, 23-25].

PD patients with LRRK2 mutations have somewhat variable pathology. Most of these cases showed the typical Lewy body pathology, however, others showed a mixture of Lewy bodies and tau inclusions, and sometimes tau inclusions alone [26]. The fact that LRRK2 mutations are associated with both α-synuclein and tau pathologies raises the possibility of LRRK2 acting upstream of α-synuclein and tau aggregation.

α-synuclein is the primary component of the Lewy bodies and Lewy neurites [27], and is normally present in the presynaptic terminals [28, 29]. At the presynaptic terminal, α-synuclein promotes the formation of the SNARE complex, regulating the neurotransmitter release [30]. α-synuclein is also known to recognize defective bilayer membranes [31] and produce membrane curvature [32]. These abilities may be related to its function in vesicle dynamics and trafficking. It is still not clear whether the normal function of α-synuclein has relevant roles in PD pathogenesis.

Recombinant α-synuclein protein can generate amyloid fibrils that are indistinguishable to the ones found in Lewy bodies [33]. All the known mutations in SNCA linked to familial PD have common molecular phenotype of accelerated aggregation of α-synuclein (reviewed in [34]). Overproduction of α-synuclein in various animal models, including mice, rats, non-human primates, flies, and nematodes, resulted in aggregation of this protein followed by cell death [35, 36]. Reduction of α-synuclein aggregates by using pharmacological agents or overexpression of aggregation inhibitors, such as α-synuclein, prevented neuronal death and behavioral deficits caused by α-synuclein [37, 38]. Collectively, these findings strongly suggest that aggregation is necessary for the mechanism by which α-synuclein exerts its pathogenic actions.

Several mechanisms and cellular targets of α-synuclein have been suggested. These include vesicle trafficking [39], microtubule-dependent transport [40], Golgi fragmentation [41], synaptic vesicle cycling [42], autophagy [43], proteasomal activity [44], and mitochondrial function [45]. It is likely that α-synuclein aggregates have multiple targets, thereby leading to various types of impairment in cellular functions.

Spreading of α-synuclein aggregates

Lewy body pathology throughout the nervous system, both the CNS and PNS, and spreads within the CNS with a very specific topographical sequence during PD progression. Braak and colleagues suggested that Lewy body and Lewy neurite pathology first appears in the olfactory bulb and the dorsal motor nucleus of vagus, followed by the propagation through the mid brain and then to the mesocortex and the neocortex. This pathological propagation may explain the symptomatic progression of the disease. Therefore, although not all patients follow the same pattern of pathological propagation due to heterogeneity of the disease, it is important to note that protein aggregate pathology can spread to larger brain regions as the disease progresses, and understanding the mechanism of the aggregate propagation would provide critical insights into how disease progression occurs. Spreading of α-synuclein aggregates in human patients has been supported by recent observations that PD patients who had received transplantation of embryonic mesencephalic tissues displayed α-synuclein-positive Lewy bodies and Lewy neurites in grafted neurons [46, 47].

Mechanism of aggregate spreading

There is an increasing body of evidence that cell-to-cell transmission of α-synuclein aggregates is the underlying mechanism for spreading of Lewy pathology [48]. Our previous studies suggested that α-synuclein aggregates are released from neurons through the unconventional exocytosis [49]. Some suggested exosome-associated secretion [50], and others proposed exophagy of α-synuclein, a mechanism involves fusion of the autophagosomes and the plasma membrane [51]. Secretion of α-synuclein appears to be an ongoing process even in healthy neurons [49], however, when neurons were under stress conditions, such as oxidative stress and failure in protein quality control systems, vesicle translocation and secretion of α-synuclein were increased [49, 50, 52, 53].

Secreted α-synuclein aggregates are transferred to neighboring neurons through endocytosis [48, 54]. Endocytosed aggregates undergo trafficking through the endocytic pathway and were delivered to the lysosomes and degraded [55]. Impaired lysosomal function caused accumulation of internalized exogenous α-synuclein, thereby promoting cell-to-cell transmission [48]. Therefore, efficient trafficking and clearance of exogenous α-synuclein aggregates would prevent seeded aggregation of the endogenous α-synuclein. The mechanism of cell-to-cell transmission of α-synuclein aggregates and how the transferred seed of aggregates are cleared have become critical questions in understanding the mode of disease progression.

LRRK2 is a large, multi-domain GTPase/kinase protein [56], characterized by the presence of a ROC domain, a COR domain, and a MAPKKK domain [57-59]. These domains are known to control several cellular functions (Fig. 1) such as proliferation, differentiation, and survival [60]. Importantly, LRRK2 has been shown to play a significant role in regulating neural functions [61]. Mammalian LRRK2 has been discovered to control neurite outgrowth; mutant LRRK2 expression in neurons inhibited neurite outgrowth while knockdown of LRRK2 promotes it [61]. This has been further demonstrated in C. elegans model where over-expressed WT LRRK2, R1441C, and G2019S mutants displayed neurite impairment and loss of dopaminergic neurons with more severe loss of DA neurons in R1441C and G2019S mutant expressors than the WT expressor [62]. Over-expression of WT LRRK2, R1441C, and G2019S mutants also led to age-dependent behavioral deficits. Interestingly, LRRK2 has been observed not only in brain cells, but also in other peripheral organs such as kidney, lung, and spleen [63]. And since LRRK2 expression was notably high in immune cells like macrophagy and monocytes, it is suggested that LRRK2 might play a role in immune functions [64].

Role of kinase activity

LRRK2 protein contains a kinase and a GTPase domain and these two domains are known to harbor most of PD-linked mutations. Increased kinase activity associated with several mutations, such as G2019S, I2012T, R1441G/C, and Y1699C, has been shown to be responsible for increased neurotoxicity [65,66]. G2019S is the most common mutation of LRRK2 and exerts its pathogenic action by toxic gain-of-function through increased kinase activity [67].

Recent findings suggested that the major pathology associated with LRRK2 mutations was Lewy bodies, although some cases showed mixture of Lewy body and tau pathology, and rarely tau-only pathology [68,69]. However, the relationship between LRRK2 mutations and α-synuclein aggregation is not clearly understood. For example, it is not yet clear if LRRK2 directly phosphorylates SNCA [70]. One report showed that LRRK2 G2019S mutation may promote α-synuclein phosphorylation at S129 and increase α-synuclein aggregation [71]. However, other studies failed to show the relationship between LRRK2 G2019S mutation and increased levels of α-synuclein phosphorylation [72].

Some studies suggested that GTP binding in LRRK2 promoted the kinase activity [73, 74], and GTP-binding domain might be important for cytotoxicity of LRRK2 [75]. However, it is not quite clear how GTP binding and GTP hydrolysis regulate the kinase activity of LRRK2 and cause cytotoxicity.

Vesicle trafficking

Vesicle trafficking is among a number of pathways affected by LRRK2-dependent neuronal damage. Rab5b, one of the proteins known to interact with LRRK2 in regulating vesicle transport, resides in several different vesicle compartments and participates in membrane trafficking [76, 77]. LRRK2 has been shown to interact with Rab5b, and defects in synaptic vesicle endocytosis caused by either overexpression or knockdown of LRRK2 expression were rescued by expression of Rab5b in primary neurons [78]. Synaptic dysfunction is an early phenomenon presented in the pathogenesis of PD, and it was also reported that LRRK2 controls synaptic morphogenesis in Drosophila model [79]. Reduced LRRK2 expression led to synaptic overgrowth, while overexpression of WT LRRK2 resulted in synaptic depletion. Another study showed that LRRK2 had a significant effect on synaptic vesicle endocytosis by enhancing Endophilin A phosphorylation at S75 [80]. Perhaps, the most strong evidence for the involvement of LRRK2 in vesicle trafficking was provided by the study of MacLeod et al. [61]. This study showed that LRRK2 interacted with Rab7L1 (PARK16), and Rab7L deficiency resulted in neurodegeneration similar to the phenotype of LRRK2 mutant expression, whereas LRRK2-induced neurodegeneration was rescued by expression of Rab7L1. This study also showed defects of endolysosomal and Golgi-associated sorting and VPS35 defects in retromer complex by PD-associated defects in LRRK2 and Rab7L1. These defects were rescued by expression of WT VPS35. Therefore, LRRK2 collaborates with Rab7L1 and VPS35 in the endolysosomal pathway and the trafficking pathway that utilizes retromer complex, and defects in this system increase PD risk.

Using tetracycline-inducible transgenic mice, Lin et al. [81] reported that overexpression of WT LRRK2 in A53T transgenic mice caused more rapid and robust neuropathological changes, including neurodegeneration, somatic α-synuclein accumulation, astrogliosis, and microglial activation, than the A53T single transgenic mice. The deteriorating effects of LRRK2 were expression level-dependent, and expression of the PD-linked G2019S LRRK2 resulted in even more severe neuropathology that of WT LRRK2 in the A53T mice. In contrast, neuropathological changes produced by transgenic expression of A53T α-synuclein were significantly reduced in LRRK2 knockout mice. This study clearly showed the pathogenic interplay between LRRK2 and α-synuclein. Since the mice expressing LRRK2 G2019S alone did not display neuropathological changes, LRRK2 may indirectly contribute to the disease progression. However, the role of LRRK2 in synucleinopathy has been challenged by other studies. These studies failed to show changes in α-synuclein and glial pathology, neurodegeneration, behavioral deficits, and timing of premature death in LRRK2 G2019S/α-synuclein A53T double transgenic mice, compared to the A53T single transgenic mice, casting a doubt in the role of LRRK2 mutations in α-synuclein-driven pathogenesis [7, 82].

Although mouse studies have produced mixed results, human pathology still suggests the role, albeit indirect, of LRRK2 in synucleinopathy. In this section, we propose a number of possible mechanisms by which LRRK2 plays roles in modulating synucleinopathy (Fig. 2).

First, LRRK2 mutations may cause defects in protein degradation system, thereby leading to accumulation of α-synuclein. Homozygous deletion of LRRK2 gene resulted in reduced autophagy and age-dependent accumulation of α-synuclein and ubiquitinated proteins in mice [83]. LRRK2 WT and mutant proteins may also cause deficits in the degradation of α-synuclein by interfering with the chaperone-mediated autophagy [84].

Second, mutant LRRK2 expression caused proinflammatory responses from microglia [85]. Inhibition of LRRK2 kinase activity and knockdown of its expression resulted in attenuation of toll-like receptor 4-mediated microglia activation [86]. Considering that α-synuclein aggregation is sensitive to inflammatory environment [87], LRRK2 may modify synucleinopathy by altering inflammatory microenvironment surrounding neurons.

Finally, the most exciting possibility is the potential role of LRRK2 in cell-to-cell transmission of synucleinopathy. Several studies reported the role of LRRK2 in vesicle transport and autophagy (see above), which may be intimately related with the trafficking pathways of α-synuclein at multiple sites during cell-to-cell transmission. Secretion of α-synuclein is mediated by unconventional exocytosis, one possible mechanism is exophagy. LRRK2 might control α-synuclein secretion by modulating the autophagy pathway. After the transfer to the recipient cells, α-synuclein is transported through the endolysosomal pathway, where LRRK2 may control the rate of degradation of transferred α-synuclein by modulating the trafficking pathways. By either promoting or inhibiting the encounter of the exogenous and endogenous α-synuclein proteins within the endosomal system, LRRK2 may also regulate seeded aggregation of α-synuclein. Subsequent secretion of the seeded aggregates may be another site of control by LRRK2.

The possible roles of LRRK2 in development and progression of synucleinopathy described here are only speculations. However, investigations into these hypothetical mechanisms are likely to provide answers as to whether these two genes indeed interplay in generating synucleinopathy and potential targets for therapy for Lewy body diseases.

  1. Hoehn MM, Yahr MD. Parkinsonism: onset, progression and mortality. Neurology 1967;17:427-442.
    Pubmed
  2. Dauer W, Przedborski S. Parkinson's disease: mechanisms and models. Neuron 2003;39:889-909.
    Pubmed
  3. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature 1997;388:839-840.
    Pubmed
  4. Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 2003;24:197-211.
    Pubmed
  5. Lesage S, Dürr A, Tazir M, Lohmann E, Leutenegger AL, Janin S, Pollak P, Brice A, French Parkinson's Disease Genetics Study Group. LRRK2 G2019S as a cause of Parkinson's disease in North African Arabs. N Engl J Med 2006;354:422-423.
    Pubmed
  6. Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan-Ruiz C, Lichtner P, Scholz SW, Hernandez DG, Krüger R, Federoff M, Klein C, Goate A, Perlmutter J, Bonin M, Nalls MA, Illig T, Gieger C, Houlden H, Steffens M, Okun MS, Racette BA, Cookson MR, Foote KD, Fernandez HH, Traynor BJ, Schreiber S, Arepalli S, Zonozi R, Gwinn K, van der Brug M, Lopez G, Chanock SJ, Schatzkin A, Park Y, Hollenbeck A, Gao J, Huang X, Wood NW, Lorenz D, Deuschl G, Chen H, Riess O, Hardy JA, Singleton AB, Gasser T. Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nat Genet 2009;41:1308-1312.
    Pubmed
  7. Daher JP, Pletnikova O, Biskup S, Musso A, Gellhaar S, Galter D, Troncoso JC, Lee MK, Dawson TM, Dawson VL, Moore DJ. Neurodegenerative phenotypes in an A53T alpha-synuclein transgenic mouse model are independent of LRRK2. Hum Mol Genet 2012;21:2420-2431.
    Pubmed
  8. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997;276:2045-2047.
    Pubmed
  9. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, Przuntek H, Epplen JT, Schöls L, Riess O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet 1998;18:106-108.
    Pubmed
  10. Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atarés B, Llorens V, Gomez Tortosa E, del Ser T, Muñoz DG, de Yebenes JG. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004;55:164-173.
    Pubmed
  11. Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destée A. Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 2004;364:1167-1169.
    Pubmed
  12. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K. alpha-Synuclein locus triplication causes Parkinson's disease. Science 2003;302:841.
    Pubmed
  13. 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;51:296-301.
    Pubmed
  14. Paisán-Ruíz C, Jain S, Evans EW, Gilks WP, Simón J, van der Brug M, López de, Aparicio S, Gil AM, Khan N, Johnson J, Martinez JR, Nicholl D, Carrera IM, Pena AS, de Silva R, Lees A, Martí-Massó JF, Pérez-Tur J, Wood NW, Singleton AB. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron 2004;44:595-600.
    Pubmed
  15. Zimprich A, Müller-Myhsok B, Farrer M, Leitner P, Sharma M, Hulihan M, Lockhart P, Strongosky A, Kachergus J, Calne DB, Stoessl J, Uitti RJ, Pfeiffer RF, Trenkwalder C, Homann N, Ott E, Wenzel K, Asmus F, Hardy J, Wszolek Z, Gasser T. The PARK8 locus in autosomal dominant parkinsonism: confirmation of linkage and further delineation of the disease-containing interval. Am J Hum Genet 2004;74:11-19.
    Pubmed
  16. Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet 2009;18:R48-R59.
    Pubmed
  17. Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, Deligtisch A, Tagliati M, Hunt AL, Klein C, Henick B, Hailpern SM, Lipton RB, Soto-Valencia J, Risch N, Bressman SB. LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J Med 2006;354:424-425.
    Pubmed
  18. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998;392:605-608.
    Pubmed
  19. Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003;299:256-259.
    Pubmed
  20. Moore DJ, Zhang L, Dawson TM, Dawson VL. A missense mutation (L166P) in DJ-1, linked to familial Parkinson's disease, confers reduced protein stability and impairs homo-oligomerization. J Neurochem 2003;87:1558-1567.
    Pubmed
  21. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science 2004;304:1158-1160.
    Pubmed
  22. Ramirez A, Heimbach A, Gründemann J, Stiller B, Hampshire D, Cid LP, Goebel I, Mubaidin AF, Wriekat AL, Roeper J, Al-Din A, Hillmer AM, Karsak M, Liss B, Woods CG, Behrens MI, Kubisch C. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet 2006;38:1184-1191.
    Pubmed
  23. Pankratz N, Wilk JB, Latourelle JC, DeStefano AL, Halter C, Pugh EW, Doheny KF, Gusella JF, Nichols WC, Foroud T, Myers RH, PSG-PROGENI and GenePD Investigators, Coordinators and Molecular Genetic Laboratories. Genomewide association study for susceptibility genes contributing to familial Parkinson disease. Hum Genet 2009;124:593-605.
    Pubmed
  24. Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, Kawaguchi T, Tsunoda T, Watanabe M, Takeda A, Tomiyama H, Nakashima K, Hasegawa K, Obata F, Yoshikawa T, Kawakami H, Sakoda S, Yamamoto M, Hattori N, Murata M, Nakamura Y, Toda T. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease. Nat Genet 2009;41:1303-1307.
    Pubmed
  25. Edwards TL, Scott WK, Almonte C, Burt A, Powell EH, Beecham GW, Wang L, Züchner S, Konidari I, Wang G, Singer C, Nahab F, Scott B, Stajich JM, Pericak-Vance M, Haines J, Vance JM, Martin ER. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann Hum Genet 2010;74:97-109.
    Pubmed
  26. Wider C, Dickson DW, Wszolek ZK. Leucine-rich repeat kinase 2 gene-associated disease: redefining genotype-phenotype correlation. Neurodegener Dis 2010;7:175-179.
    Pubmed
  27. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson's disease and dementia with lewy bodies. Proc Natl Acad Sci U S A 1998;95:6469-6473.
    Pubmed
  28. Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, Kittel A, Saitoh T. The precursor protein of non-A beta component of Alzheimer's disease amyloid is a presynaptic protein of the central nervous system. Neuron 1995;14:467-475.
    Pubmed
  29. Maroteaux L, Campanelli JT, Scheller RH. Synuclein: a neuron-specific protein localized to the nucleus and presynaptic nerve terminal. J Neurosci 1988;8:2804-2815.
    Pubmed
  30. Burré J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Südhof TC. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010;329:1663-1667.
    Pubmed
  31. Nuscher B, Kamp F, Mehnert T, Odoy S, Haass C, Kahle PJ, Beyer K. Alpha-synuclein has a high affinity for packing defects in a bilayer membrane: a thermodynamics study. J Biol Chem 2004;279:21966-21975.
    Pubmed
  32. Varkey J, Isas JM, Mizuno N, Jensen MB, Bhatia VK, Jao CC, Petrlova J, Voss JC, Stamou DG, Steven AC, Langen R. Membrane curvature induction and tubulation are common features of synucleins and apolipoproteins. J Biol Chem 2010;285:32486-32493.
    Pubmed
  33. Conway KA, Harper JD, Lansbury PT. Fibrils formed in vitro from alpha-synuclein and two mutant forms linked to Parkinson's disease are typical amyloid. Biochemistry 2000;39:2552-2563.
    Pubmed
  34. Kim C, Lee SJ. Controlling the mass action of alpha-synuclein in Parkinson's disease. J Neurochem 2008;107:303-316.
    Pubmed
  35. Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci 2013;14:38-48.
    Pubmed
  36. Maries E, Dass B, Collier TJ, Kordower JH, Steece-Collier K. The role of alpha-synuclein in Parkinson's disease: insights from animal models. Nat Rev Neurosci 2003;4:727-738.
    Pubmed
  37. Hashimoto M, Rockenstein E, Mante M, Mallory M, Masliah E. beta-Synuclein inhibits alpha-synuclein aggregation: a possible role as an anti-parkinsonian factor. Neuron 2001;32:213-223.
    Pubmed
  38. Sarkar S, Perlstein EO, Imarisio S, Pineau S, Cordenier A, Maglathlin RL, Webster JA, Lewis TA, O'Kane CJ, Schreiber SL, Rubinsztein DC. Small molecules enhance autophagy and reduce toxicity in Huntington's disease models. Nat Chem Biol 2007;3:331-338.
    Pubmed
  39. Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, Labaer J, Rochet JC, Bonini NM, Lindquist S. Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science 2006;313:324-328.
    Pubmed
  40. Lee HJ, Khoshaghideh F, Lee S, Lee SJ. Impairment of microtubule-dependent trafficking by overexpression of alpha-synuclein. Eur J Neurosci 2006;24:3153-3162.
    Pubmed
  41. Gosavi N, Lee HJ, Lee JS, Patel S, Lee SJ. Golgi fragmentation occurs in the cells with prefibrillar alpha-synuclein aggregates and precedes the formation of fibrillar inclusion. J Biol Chem 2002;277:48984-48992.
    Pubmed
  42. Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, Chaudhry FA, Nicoll RA, Edwards RH. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 2010;65:66-79.
    Pubmed
  43. Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A, Gordon DE, Peden AA, Lichtenberg M, Menzies FM, Ravikumar B, Imarisio S, Brown S, O'Kane CJ, Rubinsztein DC. α-Synuclein impairs macroautophagy: implications for Parkinson's disease. J Cell Biol 2010;190:1023-1037.
    Pubmed
  44. Lindersson E, Beedholm R, Højrup P, Moos T, Gai W, Hendil KB, Jensen PH. Proteasomal inhibition by alpha-synuclein filaments and oligomers. J Biol Chem 2004;279:12924-12934.
    Pubmed
  45. Parihar MS, Parihar A, Fujita M, Hashimoto M, Ghafourifar P. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci 2008;65:1272-1284.
    Pubmed
  46. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med 2008;14:504-506.
    Pubmed
  47. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, Lashley T, Quinn NP, Rehncrona S, Björklund A, Widner H, Revesz T, Lindvall O, Brundin P. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med 2008;14:501-503.
    Pubmed
  48. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, Spencer B, Masliah E, Lee SJ. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A 2009;106:13010-13015.
    Pubmed
  49. Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci 2005;25:6016-6024.
    Pubmed
  50. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, Stefanis L, Vekrellis K. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci 2010;30:6838-6851.
    Pubmed
  51. Ejlerskov P, Rasmussen I, Nielsen TT, Bergström AL, Tohyama Y, Jensen PH, Vilhardt F. Tubulin polymerization-promoting protein (TPPP/p25α) promotes unconventional secretion of α-synuclein through exophagy by impairing autophagosome-lysosome fusion. J Biol Chem 2013;288:17313-17335.
    Pubmed
  52. Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ. Non-classical exocytosis of alpha-synuclein is sensitive to folding states and promoted under stress conditions. J Neurochem 2010;113:1263-1274.
    Pubmed
  53. Lee HJ, Baek SM, Ho DH, Suk JE, Cho ED, Lee SJ. Dopamine promotes formation and secretion of non-fibrillar alpha-synuclein oligomers. Exp Mol Med 2011;43:216-222.
    Pubmed
  54. Hansen C, Angot E, Bergström AL, Steiner JA, Pieri L, Paul G, Outeiro TF, Melki R, Kallunki P, Fog K, Li JY, Brundin P. α-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Invest 2011;121:715-725.
    Pubmed
  55. Lee HJ, Suk JE, Bae EJ, Lee JH, Paik SR, Lee SJ. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int J Biochem Cell Biol 2008;40:1835-1849.
    Pubmed
  56. Bosgraaf L, Van Haastert PJ. Roc, a Ras/GTPase domain in complex proteins. Biochim Biophys Acta 2003;1643:5-10.
    Pubmed
  57. Greggio E, Zambrano I, Kaganovich A, Beilina A, Taymans JM, Daniëls V, Lewis P, Jain S, Ding J, Syed A, Thomas KJ, Baekelandt V, Cookson MR. The Parkinson disease-associated leucine-rich repeat kinase 2 (LRRK2) is a dimer that undergoes intramolecular autophosphorylation. J Biol Chem 2008;283:16906-16914.
    Pubmed
  58. Klein CL, Rovelli G, Springer W, Schall C, Gasser T, Kahle PJ. Homo-and heterodimerization of ROCO kinases: LRRK2 kinase inhibition by the LRRK2 ROCO fragment. J Neurochem 2009;111:703-715.
    Pubmed
  59. Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA. LRRK2 in Parkinson's disease: protein domains and functional insights. Trends Neurosci 2006;29:286-293.
    Pubmed
  60. Milosevic J, Schwarz SC, Ogunlade V, Meyer AK, Storch A, Schwarz J. Emerging role of LRRK2 in human neural progenitor cell cycle progression, survival and differentiation. Mol Neurodegener 2009;4:25.
    Pubmed
  61. MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron 2006;52:587-593.
    Pubmed
  62. Yao C, El Khoury R, Wang W, Byrd TA, Pehek EA, Thacker C, Zhu X, Smith MA, Wilson-Delfosse AL, Chen SG. LRRK2-mediated neurodegeneration and dysfunction of dopaminergic neurons in a Caenorhabditis elegans model of Parkinson's disease. Neurobiol Dis 2010;40:73-81.
    Pubmed
  63. Maekawa T, Kubo M, Yokoyama I, Ohta E, Obata F. Age-dependent and cell-population-restricted LRRK2 expression in normal mouse spleen. Biochem Biophys Res Commun 2010;392:431-435.
    Pubmed
  64. Thévenet J, Pescini Gobert R, Hooft van Huijsduijnen R, Wiessner C, Sagot YJ. Regulation of LRRK2 expression points to a functional role in human monocyte maturation. PLoS One 2011;6:e21519.
    Pubmed
  65. West AB, Moore DJ, Choi C, Andrabi SA, Li X, Dikeman D, Biskup S, Zhang Z, Lim KL, Dawson VL, Dawson TM. Parkinson's disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity. Hum Mol Genet 2007;16:223-232.
    Pubmed
  66. Di Fonzo A, Tassorelli C, De Mari M, Chien HF, Ferreira J, Rohé CF, Riboldazzi G, Antonini A, Albani G, Mauro A, Marconi R, Abbruzzese G, Lopiano L, Fincati E, Guidi M, Marini P, Stocchi F, Onofrj M, Toni V, Tinazzi M, Fabbrini G, Lamberti P, Vanacore N, Meco G, Leitner P, Uitti RJ, Wszolek ZK, Gasser T, Simons EJ, Breedveld GJ, Goldwurm S, Pezzoli G, Sampaio C, Barbosa E, Martignoni E, Oostra BA, Bonifati V, Italian Parkinson's Genetics Network. Comprehensive analysis of the LRRK2 gene in sixty families with Parkinson's disease. Eur J Hum Genet 2006;14:322-331.
    Pubmed
  67. Lee BD, Shin JH, VanKampen J, Petrucelli L, West AB, Ko HS, Lee YI, Maguire-Zeiss KA, Bowers WJ, Federoff HJ, Dawson VL, Dawson TM. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson's disease. Nat Med 2010;16:998-1000.
    Pubmed
  68. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Müller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004;44:601-607.
    Pubmed
  69. Rajput A, Dickson DW, Robinson CA, Ross OA, Dächsel JC, Lincoln SJ, Cobb SA, Rajput ML, Farrer MJ. Parkinsonism, Lrrk2 G2019S, and tau neuropathology. Neurology 2006;67:1506-1508.
    Pubmed
  70. Carballo-Carbajal I, Weber-Endress S, Rovelli G, Chan D, Wolozin B, Klein CL, Patenge N, Gasser T, Kahle PJ. Leucine-rich repeat kinase 2 induces alpha-synuclein expression via the extracellular signal-regulated kinase pathway. Cell Signal 2010;22:821-827.
    Pubmed
  71. Qing H, Wong W, McGeer EG, McGeer PL. Lrrk2 phosphorylates alpha synuclein at serine 129: Parkinson disease implications. Biochem Biophys Res Commun 2009;387:149-152.
    Pubmed
  72. Liu G, Aliaga L, Cai H. α-Synuclein, LRRK2 and their interplay in Parkinson's disease. Future Neurol 2012;7:145-153.
    Pubmed
  73. Ito G, Okai T, Fujino G, Takeda K, Ichijo H, Katada T, Iwatsubo T. GTP binding is essential to the protein kinase activity of LRRK2, a causative gene product for familial Parkinson's disease. Biochemistry 2007;46:1380-1388.
    Pubmed
  74. Guo L, Gandhi PN, Wang W, Petersen RB, Wilson-Delfosse AL, Chen SG. The Parkinson's disease-associated protein, leucine-rich repeat kinase 2 (LRRK2), is an authentic GTPase that stimulates kinase activity. Exp Cell Res 2007;313:3658-3670.
    Pubmed
  75. Xiong Y, Coombes CE, Kilaru A, Li X, Gitler AD, Bowers WJ, Dawson VL, Dawson TM, Moore DJ. GTPase activity plays a key role in the pathobiology of LRRK2. PLoS Genet 2010;6:e1000902.
    Pubmed
  76. Seabra MC, Wasmeier C. Controlling the location and activation of Rab GTPases. Curr Opin Cell Biol 2004;16:451-457.
    Pubmed
  77. Pfeffer S, Aivazian D. Targeting Rab GTPases to distinct membrane compartments. Nat Rev Mol Cell Biol 2004;5:886-896.
    Pubmed
  78. Shin N, Jeong H, Kwon J, Heo HY, Kwon JJ, Yun HJ, Kim CH, Han BS, Tong Y, Shen J, Hatano T, Hattori N, Kim KS, Chang S, Seol W. LRRK2 regulates synaptic vesicle endocytosis. Exp Cell Res 2008;314:2055-2065.
    Pubmed
  79. Lee S, Liu HP, Lin WY, Guo H, Lu B. LRRK2 kinase regulates synaptic morphology through distinct substrates at the presynaptic and postsynaptic compartments of the Drosophila neuromuscular junction. J Neurosci 2010;30:16959-16969.
    Pubmed
  80. Matta S, Van Kolen K, da Cunha R, van den Bogaart G, Mandemakers W, Miskiewicz K, De Bock PJ, Morais VA, Vilain S, Haddad D, Delbroek L, Swerts J, Chávez-Gutiérrez L, Esposito G, Daneels G, Karran E, Holt M, Gevaert K, Moechars DW, De Strooper B, Verstreken P. LRRK2 controls an EndoA phosphorylation cycle in synaptic endocytosis. Neuron 2012;75:1008-1021.
    Pubmed
  81. Lin X, Parisiadou L, Gu XL, Wang L, Shim H, Sun L, Xie C, Long CX, Yang WJ, Ding J, Chen ZZ, Gallant PE, Tao-Cheng JH, Rudow G, Troncoso JC, Liu Z, Li Z, Cai H. Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson's-disease-related mutant alpha-synuclein. Neuron 2009;64:807-827.
    Pubmed
  82. Herzig MC, Bidinosti M, Schweizer T, Hafner T, Stemmelen C, Weiss A, Danner S, Vidotto N, Stauffer D, Barske C, Mayer F, Schmid P, Rovelli G, van der Putten PH, Shimshek DR. High LRRK2 levels fail to induce or exacerbate neuronal alpha-synucleinopathy in mouse brain. PLoS One 2012;7:e36581.
    Pubmed
  83. Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher RJ, Shen J. Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha-synuclein, and apoptotic cell death in aged mice. Proc Natl Acad Sci U S A 2010;107:9879-9884.
    Pubmed
  84. Orenstein SJ, Kuo SH, Tasset I, Arias E, Koga H, Fernandez-Carasa I, Cortes E, Honig LS, Dauer W, Consiglio A, Raya A, Sulzer D, Cuervo AM. Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci 2013;16:394-406.
    Pubmed
  85. Gillardon F, Schmid R, Draheim H. Parkinson's disease-linked leucine-rich repeat kinase 2(R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity. Neuroscience 2012;208:41-48.
    Pubmed
  86. Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, Cowell RM, West AB. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci 2012;32:1602-1611.
    Pubmed
  87. Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci 2008;28:7687-7698.
    Pubmed