Skip to main content
Log in

Recent Developments in LRRK2-Targeted Therapy for Parkinson’s Disease

  • Leading Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Kinase activating missense mutations in leucine-rich repeat kinase 2 (LRRK2) are pathogenically linked to neurodegenerative Parkinson’s disease (PD). Over the past decade, substantial effort has been devoted to the development of potent and selective small molecule inhibitors of LRRK2, as well as their preclinical testing across different Parkinson’s disease models. This review outlines the genetic and biochemical evidence that pathogenic missense mutations increase LRRK2 kinase activity, which in turn provides the rationale for the development of small molecule inhibitors as potential PD therapeutics. An overview of progress in the development of LRRK2 inhibitors is provided, which in particular indicates that highly selective and potent compounds capable of clinical utility have been developed. We outline evidence from rodent- and human-induced pluripotent stem cell models that support a pathogenic role for LRRK2 kinase activity, and review the substantial experiments aimed at evaluating the safety of LRRK2 inhibitors. We address challenges still to overcome in the translational therapeutic pipeline, including biomarker development and clinical trial strategies, and finally outline the potential utility of LRRK2 inhibitors for other genetic forms of PD and ultimately sporadic PD. Collective evidence supports the ongoing clinical translation of LRRK2 inhibitors as a therapeutic intervention for PD is greatly needed.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39(6):889–909.

    CAS  PubMed  Google Scholar 

  2. Ross GW, Petrovitch H, Abbott RD, Nelson J, Markesbery W, Davis D, et al. Parkinsonian signs and substantia nigra neuron density in decendents elders without PD. Ann Neurol. 2004;56(4):532–9.

    PubMed  Google Scholar 

  3. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386(9996):896–912.

    CAS  PubMed  Google Scholar 

  4. Chaudhuri KR, Healy DG, Schapira AH. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol. 2006;5(3):235–45.

    PubMed  Google Scholar 

  5. Halliday G, Lees A, Stern M. Milestones in Parkinson’s disease—clinical and pathologic features. Mov Disord. 2011;26(6):1015–21.

    PubMed  Google Scholar 

  6. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.

    CAS  PubMed  Google Scholar 

  7. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211.

    PubMed  Google Scholar 

  8. Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 2008;116(1):1–16.

    CAS  PubMed  Google Scholar 

  9. Parkkinen L, Pirttila T, Alafuzoff I. Applicability of current staging/categorization of alpha-synuclein pathology and their clinical relevance. Acta Neuropathol. 2008;115(4):399–407.

    PubMed  PubMed Central  Google Scholar 

  10. Gasser T, Hardy J, Mizuno Y. Milestones in PD genetics. Mov Disord. 2011;26(6):1042–8.

    PubMed  Google Scholar 

  11. Houlden H, Singleton AB. The genetics and neuropathology of Parkinson’s disease. Acta Neuropathol. 2012;124(3):325–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Lill CM. Genetics of Parkinson’s disease. Mol Cell Probes. 2016;30(6):386–96.

    CAS  PubMed  Google Scholar 

  13. Trinh J, Farrer M. Advances in the genetics of Parkinson disease. Nat Rev Neurol. 2013;9(8):445–54.

    CAS  PubMed  Google Scholar 

  14. Paisan-Ruiz C, Jain S, Evans EW, Gilks WP, Simon J, van der Brug M, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron. 2004;44(4):595–600.

    CAS  PubMed  Google Scholar 

  15. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44(4):601–7.

    CAS  PubMed  Google Scholar 

  16. Domingo A, Klein C. Genetics of Parkinson disease. Handb Clin Neurol. 2018;147:211–27.

    PubMed  Google Scholar 

  17. Nuytemans K, Theuns J, Cruts M, Van Broeckhoven C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat. 2010;31(7):763–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Paisan-Ruiz C, Lewis PA, Singleton AB. LRRK2: cause, risk, and mechanism. J Parkinsons Dis. 2013;3(2):85–103.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Healy DG, Falchi M, O’Sullivan SS, Bonifati V, Durr A, Bressman S, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study. Lancet Neurol. 2008;7(7):583–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Haugarvoll K, Rademakers R, Kachergus JM, Nuytemans K, Ross OA, Gibson JM, et al. Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease. Neurology. 2008;70(16 Pt 2):1456–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Atashrazm F, Dzamko N. LRRK2 inhibitors and their potential in the treatment of Parkinson’s disease: current perspectives. Clin Pharmacol. 2016;8:177–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Cookson MR. LRRK2 pathways leading to neurodegeneration. Curr Neurol Neurosci Rep. 2015;15(7):42.

    PubMed  PubMed Central  Google Scholar 

  23. Harvey K, Outeiro TF. The role of LRRK2 in cell signalling. Biochem Soc Trans. 2019;47(1):23–44.

    Google Scholar 

  24. Monfrini E, Di Fonzo A. Leucine-rich repeat kinase (LRRK2) genetics and Parkinson’s disease. Adv Neurobiol. 2017;14:3–30.

    PubMed  Google Scholar 

  25. Hernandez DG, Reed X, Singleton AB. Genetics in Parkinson disease: mendelian versus non-mendelian inheritance. J Neurochem. 2016;139(Suppl 1):59–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cookson MR. The role of leucine-rich repeat kinase 2 (LRRK2) in Parkinson’s disease. Nat Rev Neurosci. 2010;11(12):791–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Lesage S, Durr A, Tazir M, Lohmann E, Leutenegger AL, Janin S, et al. LRRK2 G2019S as a cause of Parkinson’s disease in North African Arabs. N Engl J Med. 2006;354(4):422–3.

    CAS  PubMed  Google Scholar 

  28. Gorostidi A, Ruiz-Martinez J, de Munain LA, Alzualde A, Marti Masso JF. LRRK2 G2019S and R1441G mutations associated with Parkinson’s disease are common in the Basque Country, but relative prevalence is determined by ethnicity. Neurogenetics. 2009;10(2):157–9.

    CAS  PubMed  Google Scholar 

  29. Wszolek ZK, Pfeiffer RF, Tsuboi Y, Uitti RJ, McComb RD, Stoessl AJ, et al. Autosomal dominant parkinsonism associated with variable synuclein and tau pathology. Neurology. 2004;62(9):1619–22.

    CAS  PubMed  Google Scholar 

  30. Berg D, Schweitzer KJ, Leitner P, Zimprich A, Lichtner P, Belcredi P, et al. Type and frequency of mutations in the LRRK2 gene in familial and sporadic Parkinson’s disease. Brain. 2005;128(Pt 12):3000–11.

    PubMed  Google Scholar 

  31. Hasegawa K, Stoessl AJ, Yokoyama T, Kowa H, Wszolek ZK, Yagishita S. Familial parkinsonism: study of original Sagamihara PARK8 (I2020T) kindred with variable clinicopathologic outcomes. Parkinsonism Relat Disord. 2009;15(4):300–6.

    PubMed  Google Scholar 

  32. Ujiie S, Hatano T, Kubo S, Imai S, Sato S, Uchihara T, et al. LRRK2 I2020T mutation is associated with tau pathology. Parkinsonism Relat Disord. 2012;18(7):819–23.

    PubMed  Google Scholar 

  33. Jaleel M, Nichols RJ, Deak M, Campbell DG, Gillardon F, Knebel A, et al. LRRK2 phosphorylates moesin at threonine-558: characterization of how Parkinson’s disease mutants affect kinase activity. Biochem J. 2007;405(2):307–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. West AB, Moore DJ, Biskup S, Bugayenko A, Smith WW, Ross CA, et al. Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci USA. 2005;102(46):16842–7.

    CAS  PubMed  Google Scholar 

  35. Sheng Z, Zhang S, Bustos D, Kleinheinz T, Le Pichon CE, Dominguez SL, et al. Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations. Sci Transl Med. 2012;4(164):164ra1.

    Google Scholar 

  36. Thirstrup K, Dachsel JC, Oppermann FS, Williamson DS, Smith GP, Fog K, et al. Selective LRRK2 kinase inhibition reduces phosphorylation of endogenous Rab10 and Rab12 in human peripheral mononuclear blood cells. Sci Rep. 2017;7(1):10300.

    PubMed  PubMed Central  Google Scholar 

  37. Steger M, Tonelli F, Ito G, Davies P, Trost M, Vetter M, et al. Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. Elife. 2016. https://doi.org/10.7554/elife.12813.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009;10(8):513–25.

    CAS  PubMed  Google Scholar 

  39. Steger M, Diez F, Dhekne HS, Lis P, Nirujogi RS, Karayel O, et al. Systematic proteomic analysis of LRRK2-mediated Rab GTPase phosphorylation establishes a connection to ciliogenesis. Elife. 2017. https://doi.org/10.7554/elife.31012.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Ito G, Katsemonova K, Tonelli F, Lis P, Baptista MA, Shpiro N, et al. Phos-tag analysis of Rab10 phosphorylation by LRRK2: a powerful assay for assessing kinase function and inhibitors. Biochem J. 2016;473(17):2671–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Liu Z, Bryant N, Kumaran R, Beilina A, Abeliovich A, Cookson MR, et al. LRRK2 phosphorylates membrane-bound Rabs and is activated by GTP-bound Rab7L1 to promote recruitment to the trans-Golgi network. Hum Mol Genet. 2018;27(2):385–95.

    CAS  PubMed  Google Scholar 

  42. Purlyte E, Dhekne HS, Sarhan AR, Gomez R, Lis P, Wightman M, et al. Rab29 activation of the Parkinson’s disease-associated LRRK2 kinase. EMBO J. 2018;37(1):1–18.

    CAS  PubMed  Google Scholar 

  43. Rudenko IN, Kaganovich A, Hauser DN, Beylina A, Chia R, Ding J, et al. The G2385R variant of leucine-rich repeat kinase 2 associated with Parkinson’s disease is a partial loss-of-function mutation. Biochem J. 2012;446(1):99–111.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Daniel G, Moore DJ. Modeling LRRK2 pathobiology in Parkinson’s disease: from yeast to rodents. Curr Top Behav Neurosci. 2015;22:331–68.

    CAS  PubMed  Google Scholar 

  45. Martin I, Kim JW, Dawson VL, Dawson TM. LRRK2 pathobiology in Parkinson’s disease. J Neurochem. 2014;131(5):554–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Cookson MR. Mechanisms of mutant LRRK2 neurodegeneration. Adv Neurobiol. 2017;14:227–39.

    PubMed  Google Scholar 

  47. Lee BD, Shin JH, VanKampen J, Petrucelli L, West AB, Ko HS, et al. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease. Nat Med. 2010;16(9):998–1000.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Xiong Y, Neifert S, Karuppagounder SS, Liu Q, Stankowski JN, Lee BD, et al. Robust kinase- and age-dependent dopaminergic and norepinephrine neurodegeneration in LRRK2 G2019S transgenic mice. Proc Natl Acad Sci USA. 2018;115(7):1635–40.

    CAS  PubMed  Google Scholar 

  49. Daher JP, Abdelmotilib HA, Hu X, Volpicelli-Daley LA, Moehle MS, Fraser KB, et al. Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates alpha-synuclein gene-induced neurodegeneration. J Biol Chem. 2015;290(32):19433–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Bae EJ, Kim DK, Kim C, Mante M, Adame A, Rockenstein E, et al. LRRK2 kinase regulates alpha-synuclein propagation via RAB35 phosphorylation. Nat Commun. 2018;9(1):3465.

    PubMed  PubMed Central  Google Scholar 

  51. Volpicelli-Daley LA, Abdelmotilib H, Liu Z, Stoyka L, Daher JP, Milnerwood AJ, et al. G2019S-LRRK2 expression augments alpha-synuclein sequestration into inclusions in neurons. J Neurosci. 2016;36(28):7415–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lavalley NJ, Slone SR, Ding H, West AB, Yacoubian TA. 14-3-3 Proteins regulate mutant LRRK2 kinase activity and neurite shortening. Hum Mol Genet. 2016;25(1):109–22.

    CAS  PubMed  Google Scholar 

  53. Qin Q, Zhi LT, Li XT, Yue ZY, Li GZ, Zhang H. Effects of LRRK2 inhibitors on nigrostriatal dopaminergic neurotransmission. CNS Neurosci Ther. 2017;23(2):162–73.

    CAS  PubMed  Google Scholar 

  54. Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, et al. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32(5):1602–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Longo F, Mercatelli D, Novello S, Arcuri L, Brugnoli A, Vincenzi F, et al. Age-dependent dopamine transporter dysfunction and Serine129 phospho-alpha-synuclein overload in G2019S LRRK2 mice. Acta Neuropathol Commun. 2017;5(1):22.

    PubMed  PubMed Central  Google Scholar 

  56. Giesert F, Glasl L, Zimprich A, Ernst L, Piccoli G, Stautner C, et al. The pathogenic LRRK2 R1441C mutation induces specific deficits modeling the prodromal phase of Parkinson’s disease in the mouse. Neurobiol Dis. 2017;105:179–93.

    CAS  PubMed  Google Scholar 

  57. Novello S, Arcuri L, Dovero S, Dutheil N, Shimshek DR, Bezard E, Morari M. G2019S LRRK2 mutation facilitates alpha-synuclein neuropathology in aged mice. Neurobiol Dis. 2018;120:21–33.

    CAS  PubMed  Google Scholar 

  58. Volta M, Beccano-Kelly DA, Paschall SA, Cataldi S, MacIsaac SE, Kuhlmann N, et al. Initial elevations in glutamate and dopamine neurotransmission decline with age, as does exploratory behavior, in LRRK2 G2019S knock-in mice. Elife. 2017;6:e28377.

    PubMed  PubMed Central  Google Scholar 

  59. Weykopf B, Haupt S, Jungverdoben J, Flitsch LJ, Hebisch M, Liu GH, et al. Induced pluripotent stem cell-based modeling of mutant LRRK2-associated Parkinson’s disease. Eur J Neurosci. 2019;49(4):561–89.

    PubMed  Google Scholar 

  60. Liu GH, Qu J, Suzuki K, Nivet E, Li M, Montserrat N, et al. Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature. 2012;491(7425):603–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Howlett EH, Jensen N, Belmonte F, Zafar F, Hu X, Kluss J, et al. LRRK2 G2019S-induced mitochondrial DNA damage is LRRK2 kinase dependent and inhibition restores mtDNA integrity in Parkinson’s disease. Hum Mol Genet. 2017;26(22):4340–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Cooper O, Seo H, Andrabi S, Guardia-Laguarta C, Graziotto J, Sundberg M, et al. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci Transl Med. 2012;4(141):141ra90.

    PubMed  PubMed Central  Google Scholar 

  63. Schwab AJ, Ebert AD. Neurite aggregation and calcium dysfunction in iPSC-derived sensory neurons with parkinson’s disease-related LRRK2 G2019S mutation. Stem Cell Rep. 2015;5(6):1039–52.

    CAS  Google Scholar 

  64. West AB. Achieving neuroprotection with LRRK2 kinase inhibitors in Parkinson disease. Exp Neurol. 2017;298(Pt B):236–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Christensen KV, Smith GP, Williamson DS. Development of LRRK2 inhibitors for the treatment of parkinson’s disease. Prog Med Chem. 2017;56:37–80.

    CAS  PubMed  Google Scholar 

  66. Hatcher JM, Choi HG, Alessi DR, Gray NS. Small-molecule inhibitors of LRRK2. Adv Neurobiol. 2017;14:241–64.

    PubMed  Google Scholar 

  67. Galatsis P. Leucine-rich repeat kinase 2 inhibitors: a patent review (2014–2016). Expert Opin Ther Pat. 2017;27(6):667–76.

    CAS  PubMed  Google Scholar 

  68. Nichols RJ, Dzamko N, Hutti JE, Cantley LC, Deak M, Moran J, et al. Substrate specificity and inhibitors of LRRK2, a protein kinase mutated in Parkinson’s disease. Biochem J. 2009;424(1):47–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Covy JP, Giasson BI. Identification of compounds that inhibit the kinase activity of leucine-rich repeat kinase 2. Biochem Biophys Res Commun. 2009;378(3):473–7.

    CAS  PubMed  Google Scholar 

  70. Anand VS, Reichling LJ, Lipinski K, Stochaj W, Duan W, Kelleher K, et al. Investigation of leucine-rich repeat kinase 2: enzymological properties and novel assays. FEBS J. 2009;276(2):466–78.

    CAS  PubMed  Google Scholar 

  71. Deng X, Dzamko N, Prescott A, Davies P, Liu Q, Yang Q, et al. Characterization of a selective inhibitor of the Parkinson’s disease kinase LRRK2. Nat Chem Biol. 2011;7(4):203–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Ramsden N, Perrin J, Ren Z, Lee BD, Zinn N, Dawson VL, et al. Chemoproteomics-based design of potent LRRK2-selective lead compounds that attenuate Parkinson’s disease-related toxicity in human neurons. ACS Chem Biol. 2011;6(10):1021–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Yao C, Johnson WM, Gao Y, Wang W, Zhang J, Deak M, et al. Kinase inhibitors arrest neurodegeneration in cell and C. elegans models of LRRK2 toxicity. Hum Mol Genet. 2013;22(2):328–44.

    CAS  PubMed  Google Scholar 

  74. Choi HG, Zhang J, Deng X, Hatcher JM, Patricelli MP, Zhao Z, et al. Brain penetrant LRRK2 inhibitor. ACS Med Chem Lett. 2012;3(8):658–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Hatcher JM, Zhang J, Choi HG, Ito G, Alessi DR, Gray NS. Discovery of a pyrrolopyrimidine (JH-II-127), a highly potent, selective, and brain penetrant LRRK2 inhibitor. ACS Med Chem Lett. 2015;6(5):584–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Reith AD, Bamborough P, Jandu K, Andreotti D, Mensah L, Dossang P, et al. GSK2578215A; a potent and highly selective 2-arylmethyloxy-5-substitutent-N-arylbenzamide LRRK2 kinase inhibitor. Bioorg Med Chem Lett. 2012;22(17):5625–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Estrada AA, Chan BK, Baker-Glenn C, Beresford A, Burdick DJ, Chambers M, et al. Discovery of highly potent, selective, and brain-penetrant aminopyrazole leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors. J Med Chem. 2014;57(3):921–36.

    CAS  PubMed  Google Scholar 

  78. Estrada AA, Liu X, Baker-Glenn C, Beresford A, Burdick DJ, Chambers M, et al. Discovery of highly potent, selective, and brain-penetrable leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors. J Med Chem. 2012;55(22):9416–33.

    CAS  PubMed  Google Scholar 

  79. Henderson JL, Kormos BL, Hayward MM, Coffman KJ, Jasti J, Kurumbail RG, et al. Discovery and preclinical profiling of 3-[4-(morpholin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-5-yl]benzonitrile (PF-06447475), a highly potent, selective, brain penetrant, and in vivo active LRRK2 kinase inhibitor. J Med Chem. 2015;58(1):419–32.

    CAS  PubMed  Google Scholar 

  80. Fell MJ, Mirescu C, Basu K, Cheewatrakoolpong B, DeMong DE, Ellis JM, et al. MLi-2, a potent, selective, and centrally active compound for exploring the therapeutic potential and safety of LRRK2 kinase inhibition. J Pharmacol Exp Ther. 2015;355(3):397–409.

    CAS  PubMed  Google Scholar 

  81. Scott JD, DeMong DE, Greshock TJ, Basu K, Dai X, Harris J, et al. Discovery of a 3-(4-pyrimidinyl) Indazole (MLi-2), an orally available and selective leucine-rich repeat kinase 2 (LRRK2) inhibitor that reduces brain kinase activity. J Med Chem. 2017;60(7):2983–92.

    CAS  PubMed  Google Scholar 

  82. Andersen MA, Wegener KM, Larsen S, Badolo L, Smith GP, Jeggo R, et al. PFE-360-induced LRRK2 inhibition induces reversible, non-adverse renal changes in rats. Toxicology. 2018;395:15–22.

    CAS  PubMed  Google Scholar 

  83. Baptista MA, Dave KD, Frasier MA, Sherer TB, Greeley M, Beck MJ, et al. Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS One. 2013;8(11):e80705.

    PubMed  PubMed Central  Google Scholar 

  84. Tong Y, Giaime E, Yamaguchi H, Ichimura T, Liu Y, Si H, et al. Loss of leucine-rich repeat kinase 2 causes age-dependent bi-phasic alterations of the autophagy pathway. Mol Neurodegener. 2012;7:2. https://doi.org/10.1186/1750-1326-7-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher RJ 3rd, et al. 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 USA. 2010;107(21):9879–84.

    CAS  PubMed  Google Scholar 

  86. Herzig MC, Kolly C, Persohn E, Theil D, Schweizer T, Hafner T, et al. LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum Mol Genet. 2011;20(21):4209–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Fuji RN, Flagella M, Baca M, Baptista MA, Brodbeck J, Chan BK, et al. Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci Transl Med. 2015;7(273):273ra15.

    PubMed  Google Scholar 

  88. Baptista M, Merchant K, Barret T, Bryce D, Ellis M, Estrada A, Fell M, Fiske B, et al. LRRK2 kinase inhibitors induce a reversible effect in the lungs of non-human primates with no measurable pulmonary deficits. bioRxiv. 2018. https://doi.org/10.1101/390815.

    Article  Google Scholar 

  89. Gardet A, Benita Y, Li C, Sands BE, Ballester I, Stevens C, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185(9):5577–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Hakimi M, Selvanantham T, Swinton E, Padmore RF, Tong Y, Kabbach G, et al. Parkinson’s disease-linked LRRK2 is expressed in circulating and tissue immune cells and upregulated following recognition of microbial structures. J Neural Transm. 2011;118(5):795–808.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Fan Y, Howden AJM, Sarhan AR, Lis P, Ito G, Martinez TN, et al. Interrogating Parkinson’s disease LRRK2 kinase pathway activity by assessing Rab10 phosphorylation in human neutrophils. Biochem J. 2018;475(1):23–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Hui KY, Fernandez-Hernandez H, Hu J, Schaffner A, Pankratz N, Hsu NY, et al. Functional variants in the LRRK2 gene confer shared effects on risk for Crohn’s disease and Parkinson’s disease. Sci Transl Med. 2018;10(423):eaa17795.

    Google Scholar 

  93. Liu Z, Lee J, Krummey S, Lu W, Cai H, Lenardo MJ. The kinase LRRK2 is a regulator of the transcription factor NFAT that modulates the severity of inflammatory bowel disease. Nat Immunol. 2011;12(11):1063–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Marcinek P, Jha AN, Shinde V, Sundaramoorthy A, Rajkumar R, Suryadevara NC, et al. LRRK2 and RIPK2 variants in the NOD 2-mediated signaling pathway are associated with susceptibility to Mycobacterium leprae in Indian populations. PLoS One. 2013;8(8):e73103.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhang FR, Huang W, Chen SM, Sun LD, Liu H, Li Y, et al. Genomewide association study of leprosy. N Engl J Med. 2009;361(27):2609–18.

    CAS  PubMed  Google Scholar 

  96. Dzamko NL. LRRK2 and the immune system. Adv Neurobiol. 2017;14:123–43.

    PubMed  Google Scholar 

  97. Bliederhaeuser C, Zondler L, Grozdanov V, Ruf WP, Brenner D, Melrose HL, et al. LRRK2 contributes to monocyte dysregulation in Parkinson’s disease. Acta Neuropathol Commun. 2016;4(1):123.

    PubMed  PubMed Central  Google Scholar 

  98. Speidel A, Felk S, Reinhardt P, Sterneckert J, Gillardon F. Leucine-rich repeat kinase 2 influences fate decision of human monocytes differentiated from induced pluripotent stem cells. PLoS One. 2016;11(11):e0165949.

    PubMed  PubMed Central  Google Scholar 

  99. Thevenet J, Pescini Gobert R, van Huijsduijnen HR, Wiessner C, Sagot YJ. Regulation of LRRK2 expression points to a functional role in human monocyte maturation. PLoS One. 2011;6(6):e21519.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Dzamko N, Inesta-Vaquera F, Zhang J, Xie C, Cai H, Arthur S, et al. The IkappaB kinase family phosphorylates the Parkinson’s disease kinase LRRK2 at Ser935 and Ser910 during Toll-like receptor signaling. PLoS One. 2012;7(6):e39132.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Schapansky J, Nardozzi JD, Felizia F, LaVoie MJ. Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy. Hum Mol Genet. 2014;23(16):4201–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Liu W, Liu X, Li Y, Zhao J, Liu Z, Hu Z, et al. LRRK2 promotes the activation of NLRC4 inflammasome during Salmonella Typhimurium infection. J Exp Med. 2017;214(10):3051–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Hartlova A, Herbst S, Peltier J, Rodgers A, Bilkei-Gorzo O, Fearns A, et al. LRRK2 is a negative regulator of Mycobacterium tuberculosis phagosome maturation in macrophages. EMBO J. 2018;37(12):e98694.

    PubMed  PubMed Central  Google Scholar 

  104. Dzamko N, Rowe DB, Halliday GM. Increased peripheral inflammation in asymptomatic leucine-rich repeat kinase 2 mutation carriers. Mov Disord. 2016;31(6):889–97.

    CAS  PubMed  Google Scholar 

  105. Kozina E, Sadasivan S, Jiao Y, Dou Y, Ma Z, Tan H, et al. Mutant LRRK2 mediates peripheral and central immune responses leading to neurodegeneration in vivo. Brain. 2018;141(6):1753–69.

    PubMed  Google Scholar 

  106. Denali Therapeutics Announces Positive Clinical Results From LRRK2 Inhibitor Program For Parkinson’s Disease. http://investors.denalitherapeutics.com/news-releases/news-release-details/denali-therapeutics-announces-positive-clinical-results-lrrk2-ir-pages. Denali Therapeutics Inc. 2018. Accessed 26 Feb 2019.

  107. Denali Therapeutics Announces First Patient Dosed in Phase 1b Study of DNL201 for Parkinson’s Disease. https://globenewswire.com/news-release/2018/12/10/1664447/0/en/Denali-Therapeutics-Announces-First-Patient-Dosed-in-Phase-1b-Study-of-DNL201-for-Parkinson-s-Disease.html. Denali Therapeutics Inc. 2018. Accessed 26 Feb 2019.

  108. Dzamko N, Deak M, Hentati F, Reith AD, Prescott AR, Alessi DR, et al. Inhibition of LRRK2 kinase activity leads to dephosphorylation of Ser(910)/Ser(935), disruption of 14-3-3 binding and altered cytoplasmic localization. Biochem J. 2010;430(3):405–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Nichols RJ, Dzamko N, Morrice NA, Campbell DG, Deak M, Ordureau A, et al. 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization. Biochem J. 2010;430(3):393–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Doggett EA, Zhao J, Mork CN, Hu D, Nichols RJ. Phosphorylation of LRRK2 serines 955 and 973 is disrupted by Parkinson’s disease mutations and LRRK2 pharmacological inhibition. J Neurochem. 2012;120(1):37–45.

    CAS  PubMed  Google Scholar 

  111. Perera G, Ranola M, Rowe DB, Halliday GM, Dzamko N. Inhibitor treatment of peripheral mononuclear cells from Parkinson’s disease patients further validates LRRK2 dephosphorylation as a pharmacodynamic biomarker. Sci Rep. 2016;6:31391.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Lobbestael E, Zhao J, Rudenko IN, Beylina A, Gao F, Wetter J, et al. Identification of protein phosphatase 1 as a regulator of the LRRK2 phosphorylation cycle. Biochem J. 2013;456(1):119–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Chia R, Haddock S, Beilina A, Rudenko IN, Mamais A, Kaganovich A, et al. Phosphorylation of LRRK2 by casein kinase 1alpha regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat Commun. 2014;5:5827.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Muda K, Bertinetti D, Gesellchen F, Hermann JS, von Zweydorf F, Geerlof A, et al. Parkinson-related LRRK2 mutation R1441C/G/H impairs PKA phosphorylation of LRRK2 and disrupts its interaction with 14-3-3. Proc Natl Acad Sci USA. 2014;111(1):E34–43.

    CAS  PubMed  Google Scholar 

  115. Fraser KB, Rawlins AB, Clark RG, Alcalay RN, Standaert DG, Liu N, et al. Ser(P)-1292 LRRK2 in urinary exosomes is elevated in idiopathic Parkinson’s disease. Mov Disord. 2016;31(10):1543–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Atashrazm F, Hammond D, Perera G, Bolliger MF, Matar E, Halliday GM, et al. LRRK2-mediated Rab10 phosphorylation in immune cells from Parkinson’s disease patients. Mov Disord. 2019;34(3):406–15.

    CAS  PubMed  Google Scholar 

  117. Malik N, Gifford AN, Sandell J, Tuchman D, Ding YS. Synthesis and in vitro and in vivo evaluation of [(3)H]LRRK2-IN-1 as a novel radioligand for LRRK2. Mol Imaging Biol. 2017;19(6):837–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Malik N, Tuchman D, Sandell J, Gifford A, Ding Y-S. Development of novel radioligands for imaging LRRK2 in Parkinson’s disease. J Nucl Med. 2018;59(supplement 1):1020.

    Google Scholar 

  119. Dzamko N, Gysbers AM, Bandopadhyay R, Bolliger MF, Uchino A, Zhao Y, et al. LRRK2 levels and phosphorylation in Parkinson’s disease brain and cases with restricted Lewy bodies. Mov Disord. 2017;32(3):423–32.

    CAS  PubMed  Google Scholar 

  120. Zhao Y, Perera G, Takahashi-Fujigasaki J, Mash DC, Vonsattel JPG, Uchino A, et al. Reduced LRRK2 in association with retromer dysfunction in post-mortem brain tissue from LRRK2 mutation carriers. Brain. 2018;141(2):486–95.

    PubMed  Google Scholar 

  121. DelleDonne A, Klos KJ, Fujishiro H, Ahmed Z, Parisi JE, Josephs KA, et al. Incidental Lewy body disease and preclinical Parkinson disease. Arch Neurol. 2008;65(8):1074–80.

    PubMed  Google Scholar 

  122. Latourelle JC, Sun M, Lew MF, Suchowersky O, Klein C, Golbe LI, et al. The Gly2019Ser mutation in LRRK2 is not fully penetrant in familial Parkinson’s disease: the GenePD study. BMC Med. 2008;6:32.

    PubMed  PubMed Central  Google Scholar 

  123. Marder K, Wang Y, Alcalay RN, Mejia-Santana H, Tang MX, Lee A, et al. Age-specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish LRRK2 Consortium. Neurology. 2015;85(1):89–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Mirelman A, Alcalay RN, Saunders-Pullman R, Yasinovsky K, Thaler A, Gurevich T, et al. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov Disord. 2015;30(7):981–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Mirelman A, Bernad-Elazari H, Thaler A, Giladi-Yacobi E, Gurevich T, Gana-Weisz M, et al. Arm swing as a potential new prodromal marker of Parkinson’s disease. Mov Disord. 2016;31(10):1527–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Bergareche A, Rodriguez-Oroz MC, Estanga A, Gorostidi A, de Munain LA, Castillo-Trivino T, et al. DAT imaging and clinical biomarkers in relatives at genetic risk for LRRK2 R1441G Parkinson’s disease. Mov Disord. 2016;31(3):335–43.

    PubMed  Google Scholar 

  127. Johansen KK, Wang L, Aasly JO, White LR, Matson WR, Henchcliffe C, et al. Metabolomic profiling in LRRK2-related Parkinson’s disease. PLoS One. 2009;4(10):e7551.

    PubMed  PubMed Central  Google Scholar 

  128. Vilarino-Guell C, Wider C, Ross OA, Dachsel JC, Kachergus JM, Lincoln SJ, et al. VPS35 mutations in Parkinson disease. Am J Hum Genet. 2011;89(1):162–7.

    PubMed  PubMed Central  Google Scholar 

  129. Zimprich A, Benet-Pages A, Struhal W, Graf E, Eck SH, Offman MN, et al. A mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson disease. Am J Hum Genet. 2011;89(1):168–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Seaman MN. The retromer complex—endosomal protein recycling and beyond. J Cell Sci. 2012;125(Pt 20):4693–702.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Trinh J, Zeldenrust FMJ, Huang J, Kasten M, Schaake S, Petkovic S, et al. Genotype-phenotype relations for the Parkinson’s disease genes SNCA, LRRK2, VPS35: MDSGene systematic review. Mov Disord. 2018;33(12):1857–70.

    CAS  PubMed  Google Scholar 

  132. Malik BR, Godena VK, Whitworth AJ. VPS35 pathogenic mutations confer no dominant toxicity but partial loss of function in Drosophila and genetically interact with parkin. Hum Mol Genet. 2015;24(21):6106–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Zavodszky E, Seaman MN, Moreau K, Jimenez-Sanchez M, Breusegem SY, Harbour ME, et al. Mutation in VPS35 associated with Parkinson’s disease impairs WASH complex association and inhibits autophagy. Nat Commun. 2014;5:3828.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Inoshita T, Arano T, Hosaka Y, Meng H, Umezaki Y, Kosugi S, et al. Vps35 in cooperation with LRRK2 regulates synaptic vesicle endocytosis through the endosomal pathway in Drosophila. Hum Mol Genet. 2017;26(15):2933–48.

    CAS  PubMed  Google Scholar 

  135. MacLeod DA, Rhinn H, Kuwahara T, Zolin A, Di Paolo G, McCabe BD, et al. RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson’s disease risk. Neuron. 2013;77(3):425–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Mir R, Tonelli F, Lis P, Macartney T, Polinski NK, Martinez TN, et al. The Parkinson’s disease VPS35[D620N] mutation enhances LRRK2-mediated Rab protein phosphorylation in mouse and human. Biochem J. 2018;475(11):1861–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Kilarski LL, Pearson JP, Newsway V, Majounie E, Knipe MD, Misbahuddin A, et al. Systematic review and UK-based study of PARK2 (parkin), PINK1, PARK7 (DJ-1) and LRRK2 in early-onset Parkinson’s disease. Mov Disord. 2012;27(12):1522–9.

    CAS  PubMed  Google Scholar 

  138. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011;12(1):9–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Mortiboys H, Johansen KK, Aasly JO, Bandmann O. Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2. Neurology. 2010;75(22):2017–20.

    CAS  PubMed  Google Scholar 

  140. Bonello F, Hassoun SM, Mouton-Liger F, Shin YS, Muscat A, Tesson C, et al. LRRK2 impairs PINK1/Parkin-dependent mitophagy via its kinase activity: pathologic insights into Parkinson’s disease. Hum Mol Genet. 2019. https://doi.org/10.1093/hmg/ddz004.

    Article  PubMed  Google Scholar 

  141. Hsieh CH, Shaltouki A, Gonzalez AE, da Cruz BA, Burbulla LF, St Lawrence E, et al. Functional impairment in miro degradation and mitophagy is a shared feature in familial and sporadic Parkinson’s disease. Cell Stem Cell. 2016;19(6):709–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Azkona G, de Maturana LR, Del Rio P, Sousa A, Vazquez N, Zubiarrain A, et al. LRRK2 expression is deregulated in fibroblasts and neurons from parkinson patients with mutations in PINK1. Mol Neurobiol. 2018;55(1):506–16.

    CAS  PubMed  Google Scholar 

  143. Smith GA, Jansson J, Rocha EM, Osborn T, Hallett PJ, Isacson O. Fibroblast biomarkers of sporadic parkinson’s disease and LRRK2 kinase inhibition. Mol Neurobiol. 2016;53(8):5161–77.

    CAS  PubMed  Google Scholar 

  144. Lai YC, Kondapalli C, Lehneck R, Procter JB, Dill BD, Woodroof HI, et al. Phosphoproteomic screening identifies Rab GTPases as novel downstream targets of PINK1. EMBO J. 2015;34(22):2840–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Bultron G, Kacena K, Pearson D, Boxer M, Yang R, Sathe S, et al. The risk of Parkinson’s disease in type 1 Gaucher disease. J Inherit Metab Dis. 2010;33(2):167–73.

    PubMed  PubMed Central  Google Scholar 

  146. Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R. Mutations in the glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2004;351(19):1972–7.

    CAS  PubMed  Google Scholar 

  147. Tsuji S, Choudary PV, Martin BM, Stubblefield BK, Mayor JA, Barranger JA, et al. A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher’s disease. N Engl J Med. 1987;316(10):570–5.

    CAS  PubMed  Google Scholar 

  148. Anheim M, Elbaz A, Lesage S, Durr A, Condroyer C, Viallet F, et al. Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology. 2012;78(6):417–20.

    CAS  PubMed  Google Scholar 

  149. Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009;361(17):1651–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Lerche S, Schulte C, Srulijes K, Pilotto A, Rattay TW, Hauser AK, et al. Cognitive impairment in glucocerebrosidase (GBA)-associated PD: not primarily associated with cerebrospinal fluid Abeta and Tau profiles. Mov Disord. 2017;32(12):1780–3.

    CAS  PubMed  Google Scholar 

  151. Rieckmann JC, Geiger R, Hornburg D, Wolf T, Kveler K, Jarrossay D, et al. Social network architecture of human immune cells unveiled by quantitative proteomics. Nat Immunol. 2017;18(5):583–93.

    CAS  PubMed  Google Scholar 

  152. Alcalay RN, Levy OA, Waters CC, Fahn S, Ford B, Kuo SH, et al. Glucocerebrosidase activity in Parkinson’s disease with and without GBA mutations. Brain. 2015;138(Pt 9):2648–58.

    PubMed  PubMed Central  Google Scholar 

  153. Atashrazm F, Hammond D, Perera G, Dobson-Stone C, Mueller N, Pickford R, et al. Reduced glucocerebrosidase activity in monocytes from patients with Parkinson’s disease. Sci Rep. 2018;8(1):15446.

    PubMed  PubMed Central  Google Scholar 

  154. Berger J, Lecourt S, Vanneaux V, Rapatel C, Boisgard S, Caillaud C, et al. Glucocerebrosidase deficiency dramatically impairs human bone marrow haematopoiesis in an in vitro model of Gaucher disease. Br J Haematol. 2010;150(1):93–101.

    CAS  PubMed  Google Scholar 

  155. Liu J, Halene S, Yang M, Iqbal J, Yang R, Mehal WZ, et al. Gaucher disease gene GBA functions in immune regulation. Proc Natl Acad Sci USA. 2012;109(25):10018–23.

    CAS  PubMed  Google Scholar 

  156. Mizukami H, Mi Y, Wada R, Kono M, Yamashita T, Liu Y, et al. Systemic inflammation in glucocerebrosidase-deficient mice with minimal glucosylceramide storage. J Clin Investig. 2002;109(9):1215–21.

    CAS  PubMed  Google Scholar 

  157. Henry AG, Aghamohammadzadeh S, Samaroo H, Chen Y, Mou K, Needle E, et al. Pathogenic LRRK2 mutations, through increased kinase activity, produce enlarged lysosomes with reduced degradative capacity and increase ATP13A2 expression. Hum Mol Genet. 2015;24(21):6013–28.

    CAS  PubMed  Google Scholar 

  158. Roosen DA, Cookson MR. LRRK2 at the interface of autophagosomes, endosomes and lysosomes. Mol Neurodegener. 2016;11(1):73. https://doi.org/10.1186/s13024-016-0140-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Eguchi T, Kuwahara T, Sakurai M, Komori T, Fujimoto T, Ito G, et al. LRRK2 and its substrate Rab GTPases are sequentially targeted onto stressed lysosomes and maintain their homeostasis. Proc Natl Acad Sci USA. 2018;115(39):E9115–24.

    PubMed  Google Scholar 

  160. Gan-Or Z, Dion PA, Rouleau GA. Genetic perspective on the role of the autophagy-lysosome pathway in Parkinson disease. Autophagy. 2015;11(9):1443–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Murphy KE, Gysbers AM, Abbott SK, Tayebi N, Kim WS, Sidransky E, et al. Reduced glucocerebrosidase is associated with increased alpha-synuclein in sporadic Parkinson’s disease. Brain. 2014;137(Pt 3):834–48.

    PubMed  PubMed Central  Google Scholar 

  162. Orenstein SJ, Kuo SH, Tasset I, Arias E, Koga H, Fernandez-Carasa I, et al. Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci. 2013;16(4):394–406.

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat Genet. 2009;41(12):1303–7.

    CAS  PubMed  Google Scholar 

  164. Sharma M, Ioannidis JP, Aasly JO, Annesi G, Brice A, Van Broeckhoven C, et al. Large-scale replication and heterogeneity in Parkinson disease genetic loci. Neurology. 2012;79(7):659–67.

    PubMed  PubMed Central  Google Scholar 

  165. Di Maio R, Hoffman EK, Rocha EM, Keeney MT, Sanders LH, De Miranda BR, et al. LRRK2 activation in idiopathic Parkinson’s disease. Sci Transl Med. 2018;10(451):eaar5429.

    Google Scholar 

  166. Andersen MA, Christensen KV, Badolo L, Smith GP, Jeggo R, Jensen PH, et al. Parkinson’s disease-like burst firing activity in subthalamic nucleus induced by AAV-alpha-synuclein is normalized by LRRK2 modulation. Neurobiol Dis. 2018;116:13–27.

    CAS  PubMed  Google Scholar 

  167. Daher JP, Pletnikova O, Biskup S, Musso A, Gelhaar S, Gatler 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(11):2420–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Henderson MX, Sengupta M, McGeary I, Zhang B, Olufemi MF, Brown H, Trojanowski JQ, Lee VMY. LRRK2 inhibition does not impart protection from alpha-synuclein pathology and neuron death in non-transgenic mice. Acta Neuropathol Commun. 2019;7(1):28.

    PubMed  PubMed Central  Google Scholar 

  169. Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, et al. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 2017;3:11.

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41(12):1308–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Tucci A, Nalls MA, Houlden H, Revesz T, Singleton AB, Wood NW, et al. Genetic variability at the PARK16 locus. Eur J Hum Genet. 2010;18(12):1356–9.

    PubMed  PubMed Central  Google Scholar 

  172. Beilina A, Rudenko IN, Kaganovich A, Civiero L, Chau H, Kalia SK, et al. Unbiased screen for interactors of leucine-rich repeat kinase 2 supports a common pathway for sporadic and familial Parkinson disease. Proc Natl Acad Sci USA. 2014;111(7):2626–31.

    CAS  PubMed  Google Scholar 

  173. Madero-Perez J, Fernandez B, Lara Ordonez AJ, Fdez E, Lobbestael E, Baekelandt V, et al. RAB7L1-Mediated relocalization of LRRK2 to the golgi complex causes centrosomal deficits via RAB8A. Front Mol Neurosci. 2018;11:417.

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Fujimoto T, Kuwahara T, Eguchi T, Sakurai M, Komori T, Iwatsubo T. Parkinson’s disease-associated mutant LRRK2 phosphorylates Rab7L1 and modifies trans-Golgi morphology. Biochem Biophys Res Commun. 2018;495(2):1708–15.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nicolas Dzamko.

Ethics declarations

Funding

No funding was specifically received for the publication of this review

Conflict of interest

ND receives funding for Parkinson’s disease research, including on LRRK2, from the National Health and Medical Research Council (NHMRC) (#1103757), the Michael J Fox Foundation for Parkinson’s disease research (MJFF), the Shake It Up Australia Foundation and the University of Sydney. In the past 12 months ND has received travel support from Denali Therapeutics, Neuropore therapies and MJFF and received payments for Grant reviews from the NHMRC and IRC. ND is a co-inventor and has received royalties from a patent on the use of LRRK2 phosphorylation sites as pharmacodynamic biomarkers (WO2011131980A1). YZ has no conflicts of interest to declare.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, Y., Dzamko, N. Recent Developments in LRRK2-Targeted Therapy for Parkinson’s Disease. Drugs 79, 1037–1051 (2019). https://doi.org/10.1007/s40265-019-01139-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40265-019-01139-4

Navigation