메뉴 건너뛰기




Volumn 21, Issue 6, 2017, Pages 601-610

Targeting LRRK2 in Parkinson’s disease: an update on recent developments

Author keywords

inhibitors; LRRK2; Parkinson s disease

Indexed keywords

LEUCINE RICH REPEAT KINASE 2; PHOSPHOTRANSFERASE INHIBITOR; RAB PROTEIN; ANTIPARKINSON AGENT; LRRK2 PROTEIN, HUMAN; PROTEIN KINASE INHIBITOR;

EID: 85019553951     PISSN: 14728222     EISSN: 17447631     Source Type: Journal    
DOI: 10.1080/14728222.2017.1323881     Document Type: Review
Times cited : (39)

References (105)
  • 1
    • 42049094200 scopus 로고    scopus 로고
    • Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease
    • Haugarvoll K, Rademakers R, Kachergus JM, et al. Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease. Neurology. 2008;70:1456–1460.
    • (2008) Neurology , vol.70 , pp. 1456-1460
    • Haugarvoll, K.1    Rademakers, R.2    Kachergus, J.M.3
  • 2
    • 50049104725 scopus 로고    scopus 로고
    • Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study
    • Healy DG, Falchi M, O’Sullivan SS, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease:a case-control study. Lancet Neurol. 2008;7:583–590.
    • (2008) Lancet Neurol , vol.7 , pp. 583-590
    • Healy, D.G.1    Falchi, M.2    O’Sullivan, S.S.3
  • 3
    • 84952334344 scopus 로고    scopus 로고
    • Linking a genome-wide association study signal to a LRRK2 coding variant in Parkinson’s disease
    • Foo JN, Chung SJ, Tan LC, et al. Linking a genome-wide association study signal to a LRRK2 coding variant in Parkinson’s disease. Mov Disord. 2016;31:484–487.
    • (2016) Mov Disord , vol.31 , pp. 484-487
    • Foo, J.N.1    Chung, S.J.2    Tan, L.C.3
  • 4
    • 85019650937 scopus 로고    scopus 로고
    • Genome-wide association study of Parkinson’s disease in East Asians
    • Foo JN, Tan LC, Irwan ID, et al. Genome-wide association study of Parkinson’s disease in East Asians. Hum Mol Genet. 2017;26:226–232.
    • (2017) Hum Mol Genet , vol.26 , pp. 226-232
    • Foo, J.N.1    Tan, L.C.2    Irwan, I.D.3
  • 5
    • 84946057538 scopus 로고    scopus 로고
    • Differential effect of caffeine intake in subjects with genetic susceptibility to Parkinson’s disease
    • Kumar PM, Paing SS, Li H, et al. Differential effect of caffeine intake in subjects with genetic susceptibility to Parkinson’s disease. Sci Rep. 2015;5:15492.
    • (2015) Sci Rep , vol.5 , pp. 15492
    • Kumar, P.M.1    Paing, S.S.2    Li, H.3
  • 6
    • 84947936383 scopus 로고    scopus 로고
    • Greater motor progression in patients with Parkinson disease who carry LRRK2 risk variants
    • Oosterveld LP, Allen JC, Jr., Ng EY, et al. Greater motor progression in patients with Parkinson disease who carry LRRK2 risk variants. Neurology. 2015;85:1039–1042.
    • (2015) Neurology , vol.85 , pp. 1039-1042
    • Oosterveld, L.P.1    Allen, J.C.2    Ng, E.Y.3
  • 7
    • 80052965333 scopus 로고    scopus 로고
    • Rare and common LRRK2 exonic variants in Parkinson’s disease
    • Tan EK., Rare and common LRRK2 exonic variants in Parkinson’s disease. Lancet Neurol. 2011;10:869–870.
    • (2011) Lancet Neurol , vol.10 , pp. 869-870
    • Tan, E.K.1
  • 8
    • 84887875465 scopus 로고    scopus 로고
    • Genetic testing of LRRK2 in Parkinson’s disease: is there a clinical role?
    • Buhat DM, Tan EK. Genetic testing of LRRK2 in Parkinson’s disease:is there a clinical role? Parkinsonism Relat Disord. 2014;20(Suppl 1):S54–6.
    • (2014) Parkinsonism Relat Disord , vol.20 , pp. S54-S56
    • Buhat, D.M.1    Tan, E.K.2
  • 9
    • 84949651389 scopus 로고    scopus 로고
    • Comparative blood transcriptome analysis in idiopathic and LRRK2 G2019S-associated Parkinson’s disease
    • Infante J, Prieto C, Sierra M, et al. Comparative blood transcriptome analysis in idiopathic and LRRK2 G2019S-associated Parkinson’s disease. Neurobiol Aging. 2016;38(214):e1–5.
    • (2016) Neurobiol Aging , vol.38 , Issue.214 , pp. e1-e5
    • Infante, J.1    Prieto, C.2    Sierra, M.3
  • 10
    • 84874720265 scopus 로고    scopus 로고
    • Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations
    • Sheng Z, Zhang S, Bustos D, et al. Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations. Sci Transl Med. 2012;4:164ra1.
    • (2012) Sci Transl Med , vol.4 , pp. 164ra1
    • Sheng, Z.1    Zhang, S.2    Bustos, D.3
  • 11
    • 28044460070 scopus 로고    scopus 로고
    • Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity
    • West AB, Moore DJ, Biskup S, et al. Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci U S A. 2005;102:16842–16847.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 16842-16847
    • West, A.B.1    Moore, D.J.2    Biskup, S.3
  • 12
    • 84973350290 scopus 로고    scopus 로고
    • Antioxidants inhibit neuronal toxicity in Parkinson’s disease-linked LRRK2
    • Angeles DC, Ho P, Dymock BW, et al. Antioxidants inhibit neuronal toxicity in Parkinson’s disease-linked LRRK2. Ann Clin Transl Neurol. 2016;3:288–294.
    • (2016) Ann Clin Transl Neurol , vol.3 , pp. 288-294
    • Angeles, D.C.1    Ho, P.2    Dymock, B.W.3
  • 13
    • 48249145829 scopus 로고    scopus 로고
    • Molecular biology changes associated with LRRK2 mutations in Parkinson’s disease
    • Lu YW, Tan EK. Molecular biology changes associated with LRRK2 mutations in Parkinson’s disease. J Neurosci Res. 2008;86:1895–1901.
    • (2008) J Neurosci Res , vol.86 , pp. 1895-1901
    • Lu, Y.W.1    Tan, E.K.2
  • 14
    • 84901346503 scopus 로고    scopus 로고
    • Thiol peroxidases ameliorate LRRK2 mutant-induced mitochondrial and dopaminergic neuronal degeneration in Drosophila
    • Angeles DC, Ho P, Chua LL, et al. Thiol peroxidases ameliorate LRRK2 mutant-induced mitochondrial and dopaminergic neuronal degeneration in Drosophila. Hum Mol Genet. 2014;23:3157–3165.
    • (2014) Hum Mol Genet , vol.23 , pp. 3157-3165
    • Angeles, D.C.1    Ho, P.2    Chua, L.L.3
  • 15
    • 84887945830 scopus 로고    scopus 로고
    • Targeting leucine-rich repeat kinase 2 in Parkinson’s disease
    • Chan SL, Angeles DC, Tan EK. Targeting leucine-rich repeat kinase 2 in Parkinson’s disease. Expert Opin Ther Targets. 2013;17:1471–1482.
    • (2013) Expert Opin Ther Targets , vol.17 , pp. 1471-1482
    • Chan, S.L.1    Angeles, D.C.2    Tan, E.K.3
  • 16
    • 84891776413 scopus 로고    scopus 로고
    • Mutant LRRK2 toxicity in neurons depends on LRRK2 levels and synuclein but not kinase activity or inclusion bodies
    • Skibinski G, Nakamura K, Cookson MR, et al. Mutant LRRK2 toxicity in neurons depends on LRRK2 levels and synuclein but not kinase activity or inclusion bodies. J Neurosci. 2014;34:418–433.
    • (2014) J Neurosci , vol.34 , pp. 418-433
    • Skibinski, G.1    Nakamura, K.2    Cookson, M.R.3
  • 17
    • 83855160771 scopus 로고    scopus 로고
    • Phosphorylation of LRRK2 serines 955 and 973 is disrupted by Parkinson’s disease mutations and LRRK2 pharmacological inhibition
    • Doggett EA, Zhao J, Mork CN, et al. Phosphorylation of LRRK2 serines 955 and 973 is disrupted by Parkinson’s disease mutations and LRRK2 pharmacological inhibition. J Neurochem. 2012;120:37–45.
    • (2012) J Neurochem , vol.120 , pp. 37-45
    • Doggett, E.A.1    Zhao, J.2    Mork, C.N.3
  • 18
    • 77956674229 scopus 로고    scopus 로고
    • 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization
    • Nichols RJ, Dzamko N, Morrice NA, et al. 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization. Biochem J. 2010;430:393–404.
    • (2010) Biochem J , vol.430 , pp. 393-404
    • Nichols, R.J.1    Dzamko, N.2    Morrice, N.A.3
  • 19
    • 77956655427 scopus 로고    scopus 로고
    • Inhibition of LRRK2 kinase activity leads to dephosphorylation of Ser(910)/Ser(935), disruption of 14-3-3 binding and altered cytoplasmic localization
    • Dzamko N, Deak M, Hentati F, 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;430:405–413.
    • (2010) Biochem J , vol.430 , pp. 405-413
    • Dzamko, N.1    Deak, M.2    Hentati, F.3
  • 20
    • 85006173271 scopus 로고    scopus 로고
    • LRRK2 levels and phosphorylation in Parkinson’s disease brain and cases with restricted Lewy bodies
    • Dzamko N, Gysbers AM, Bandopadhyay R, et al. LRRK2 levels and phosphorylation in Parkinson’s disease brain and cases with restricted Lewy bodies. Mov Disord. 2017;32:423–432.
    • (2017) Mov Disord , vol.32 , pp. 423-432
    • Dzamko, N.1    Gysbers, A.M.2    Bandopadhyay, R.3
  • 21
    • 0033681149 scopus 로고    scopus 로고
    • Chronic systemic pesticide exposure reproduces features of Parkinson’s disease
    • Betarbet R, Sherer TB, MacKenzie G, et al. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3:1301–1306.
    • (2000) Nat Neurosci , vol.3 , pp. 1301-1306
    • Betarbet, R.1    Sherer, T.B.2    MacKenzie, G.3
  • 22
    • 0033608811 scopus 로고    scopus 로고
    • Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss
    • Brooks AI, Chadwick CA, Gelbard HA, et al. Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 1999;823:1–10.
    • (1999) Brain Res , vol.823 , pp. 1-10
    • Brooks, A.I.1    Chadwick, C.A.2    Gelbard, H.A.3
  • 23
    • 0000428532 scopus 로고
    • 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists
    • Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature. 1957;180:1200.
    • (1957) Nature , vol.180 , pp. 1200
    • Carlsson, A.1    Lindqvist, M.2    Magnusson, T.3
  • 24
    • 0022403289 scopus 로고
    • Dopaminergic toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion after their stereotaxic administration to rats: implication for the mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity
    • Heikkila RE, Nicklas WJ, Vyas I, et al. Dopaminergic toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion after their stereotaxic administration to rats:implication for the mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity. Neurosci Lett. 1985;62:389–394.
    • (1985) Neurosci Lett , vol.62 , pp. 389-394
    • Heikkila, R.E.1    Nicklas, W.J.2    Vyas, I.3
  • 25
    • 0035091767 scopus 로고    scopus 로고
    • The parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): a technical review of its utility and safety
    • Przedborski S, Jackson-Lewis V, Naini AB, et al. The parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP):a technical review of its utility and safety. J Neurochem. 2001;76:1265–1274.
    • (2001) J Neurochem , vol.76 , pp. 1265-1274
    • Przedborski, S.1    Jackson-Lewis, V.2    Naini, A.B.3
  • 26
    • 84865535547 scopus 로고    scopus 로고
    • Use of viral vectors to create animal models for Parkinson’s disease
    • Low K, Aebischer P. Use of viral vectors to create animal models for Parkinson’s disease. Neurobiol Dis. 2012;48:189–201.
    • (2012) Neurobiol Dis , vol.48 , pp. 189-201
    • Low, K.1    Aebischer, P.2
  • 27
    • 77956441086 scopus 로고    scopus 로고
    • Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease
    • Lee BD, Shin JH, VanKampen J, et al. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease. Nat Med. 2010;16:998–1000.
    • (2010) Nat Med , vol.16 , pp. 998-1000
    • Lee, B.D.1    Shin, J.H.2    VanKampen, J.3
  • 28
    • 78751522558 scopus 로고    scopus 로고
    • A rat model of progressive nigral neurodegeneration induced by the Parkinson’s disease-associated G2019S mutation in LRRK2
    • Dusonchet J, Kochubey O, Stafa K, et al. A rat model of progressive nigral neurodegeneration induced by the Parkinson’s disease-associated G2019S mutation in LRRK2. J Neurosci. 2011;31:907–912.
    • (2011) J Neurosci , vol.31 , pp. 907-912
    • Dusonchet, J.1    Kochubey, O.2    Stafa, K.3
  • 29
    • 84924251953 scopus 로고    scopus 로고
    • Adenoviral-mediated expression of G2019S LRRK2 induces striatal pathology in a kinase-dependent manner in a rat model of Parkinson’s disease
    • Tsika E, Nguyen AP, Dusonchet J, et al. Adenoviral-mediated expression of G2019S LRRK2 induces striatal pathology in a kinase-dependent manner in a rat model of Parkinson’s disease. Neurobiol Dis. 2015;77:49–61.
    • (2015) Neurobiol Dis , vol.77 , pp. 49-61
    • Tsika, E.1    Nguyen, A.P.2    Dusonchet, J.3
  • 30
    • 84964345977 scopus 로고    scopus 로고
    • Targeting the cannabinoid CB2 receptor to attenuate the progression of motor deficits in LRRK2-transgenic mice
    • Palomo-Garo C, Gomez-Galvez Y, Garcia C, et al. Targeting the cannabinoid CB2 receptor to attenuate the progression of motor deficits in LRRK2-transgenic mice. Pharmacol Res. 2016;110:181–192.
    • (2016) Pharmacol Res , vol.110 , pp. 181-192
    • Palomo-Garo, C.1    Gomez-Galvez, Y.2    Garcia, C.3
  • 31
    • 84861552733 scopus 로고    scopus 로고
    • LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors
    • Hinkle KM, Yue M, Behrouz B, et al. LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol Neurodegener. 2012;7:25.
    • (2012) Mol Neurodegener , vol.7 , pp. 25
    • Hinkle, K.M.1    Yue, M.2    Behrouz, B.3
  • 32
    • 84879129926 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 (LRRK2)-deficient rats exhibit renal tubule injury and perturbations in metabolic and immunological homeostasis
    • Ness D, Ren Z, Gardai S, et al. Leucine-rich repeat kinase 2 (LRRK2)-deficient rats exhibit renal tubule injury and perturbations in metabolic and immunological homeostasis. Plos One. 2013;8:e66164.
    • (2013) Plos One , vol.8 , pp. e66164
    • Ness, D.1    Ren, Z.2    Gardai, S.3
  • 33
    • 84893527711 scopus 로고    scopus 로고
    • Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs
    • Baptista MA, Dave KD, Frasier MA, et al. Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. Plos One. 2013;8:e80705.
    • (2013) Plos One , vol.8 , pp. e80705
    • Baptista, M.A.1    Dave, K.D.2    Frasier, M.A.3
  • 34
    • 79952918505 scopus 로고    scopus 로고
    • Characterization of a selective inhibitor of the Parkinson’s disease kinase LRRK2
    • Deng X, Dzamko N, Prescott A, et al. Characterization of a selective inhibitor of the Parkinson’s disease kinase LRRK2. Nat Chem Biol. 2011;7:203–205.
    • (2011) Nat Chem Biol , vol.7 , pp. 203-205
    • Deng, X.1    Dzamko, N.2    Prescott, A.3
  • 35
    • 84957419608 scopus 로고    scopus 로고
    • MLi-2, a potent, selective, and centrally active compound for exploring the therapeutic potential and safety of LRRK2 kinase inhibition
    • Fell MJ, Mirescu C, Basu K, 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;355:397–409.
    • (2015) J Pharmacol Exp Ther , vol.355 , pp. 397-409
    • Fell, M.J.1    Mirescu, C.2    Basu, K.3
  • 36
    • 84939145610 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates alpha-synuclein gene-induced neurodegeneration
    • Daher JP, Abdelmotilib HA, Hu X, et al. Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates alpha-synuclein gene-induced neurodegeneration. J Biol Chem. 2015;290:19433–19444.
    • (2015) J Biol Chem , vol.290 , pp. 19433-19444
    • Daher, J.P.1    Abdelmotilib, H.A.2    Hu, X.3
  • 37
    • 36248966518 scopus 로고    scopus 로고
    • Induction of pluripotent stem cells from adult human fibroblasts by defined factors
    • Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872.
    • (2007) Cell , vol.131 , pp. 861-872
    • Takahashi, K.1    Tanabe, K.2    Ohnuki, M.3
  • 38
    • 36749043230 scopus 로고    scopus 로고
    • Induced pluripotent stem cell lines derived from human somatic cells
    • Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–1920.
    • (2007) Science , vol.318 , pp. 1917-1920
    • Yu, J.1    Vodyanik, M.A.2    Smuga-Otto, K.3
  • 39
    • 70350732790 scopus 로고    scopus 로고
    • A chemical platform for improved induction of human iPSCs
    • Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs. Nat Methods. 2009;6:805–808.
    • (2009) Nat Methods , vol.6 , pp. 805-808
    • Lin, T.1    Ambasudhan, R.2    Yuan, X.3
  • 40
    • 55849117368 scopus 로고    scopus 로고
    • Generation of mouse induced pluripotent stem cells without viral vectors
    • Okita K, Nakagawa M, Hyenjong H, et al. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322:949–953.
    • (2008) Science , vol.322 , pp. 949-953
    • Okita, K.1    Nakagawa, M.2    Hyenjong, H.3
  • 41
    • 55849115999 scopus 로고    scopus 로고
    • Induced pluripotent stem cells generated without viral integration
    • Stadtfeld M, Nagaya M, Utikal J, et al. Induced pluripotent stem cells generated without viral integration. Science. 2008;322:945–949.
    • (2008) Science , vol.322 , pp. 945-949
    • Stadtfeld, M.1    Nagaya, M.2    Utikal, J.3
  • 42
    • 77958536106 scopus 로고    scopus 로고
    • Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA
    • Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010;7:618–630.
    • (2010) Cell Stem Cell , vol.7 , pp. 618-630
    • Warren, L.1    Manos, P.D.2    Ahfeldt, T.3
  • 43
    • 62149125434 scopus 로고    scopus 로고
    • Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling
    • Chambers SM, Fasano CA, Papapetrou EP, et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–280.
    • (2009) Nat Biotechnol , vol.27 , pp. 275-280
    • Chambers, S.M.1    Fasano, C.A.2    Papapetrou, E.P.3
  • 44
    • 84862777308 scopus 로고    scopus 로고
    • Direct reprogramming of fibroblasts into neural stem cells by defined factors
    • Han DW, Tapia N, Hermann A, et al. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell. 2012;10:465–472.
    • (2012) Cell Stem Cell , vol.10 , pp. 465-472
    • Han, D.W.1    Tapia, N.2    Hermann, A.3
  • 45
    • 84859555919 scopus 로고    scopus 로고
    • Direct conversion of fibroblasts into stably expandable neural stem cells
    • Thier M, Worsdorfer P, Lakes YB, et al. Direct conversion of fibroblasts into stably expandable neural stem cells. Cell Stem Cell. 2012;10:473–479.
    • (2012) Cell Stem Cell , vol.10 , pp. 473-479
    • Thier, M.1    Worsdorfer, P.2    Lakes, Y.B.3
  • 46
    • 79952172335 scopus 로고    scopus 로고
    • LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress
    • Nguyen HN, Byers B, Cord B, et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell. 2011;8:267–280.
    • (2011) Cell Stem Cell , vol.8 , pp. 267-280
    • Nguyen, H.N.1    Byers, B.2    Cord, B.3
  • 47
    • 84887004639 scopus 로고    scopus 로고
    • Inhibition of excessive mitochondrial fission reduced aberrant autophagy and neuronal damage caused by LRRK2 G2019S mutation
    • Su YC, Qi X. Inhibition of excessive mitochondrial fission reduced aberrant autophagy and neuronal damage caused by LRRK2 G2019S mutation. Hum Mol Genet. 2013;22:4545–4561.
    • (2013) Hum Mol Genet , vol.22 , pp. 4545-4561
    • Su, Y.C.1    Qi, X.2
  • 48
    • 84875640261 scopus 로고    scopus 로고
    • Interplay of LRRK2 with chaperone-mediated autophagy
    • Orenstein SJ, Kuo SH, Tasset I, et al. Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci. 2013;16:394–406.
    • (2013) Nat Neurosci , vol.16 , pp. 394-406
    • Orenstein, S.J.1    Kuo, S.H.2    Tasset, I.3
  • 49
    • 84887506882 scopus 로고    scopus 로고
    • LRRK2 mutations cause mitochondrial DNA damage in iPSC-derived neural cells from Parkinson’s disease patients: reversal by gene correction
    • Sanders LH, Laganiere J, Cooper O, et al. LRRK2 mutations cause mitochondrial DNA damage in iPSC-derived neural cells from Parkinson’s disease patients:reversal by gene correction. Neurobiol Dis. 2014;62:381–386.
    • (2014) Neurobiol Dis , vol.62 , pp. 381-386
    • Sanders, L.H.1    Laganiere, J.2    Cooper, O.3
  • 50
    • 84949513552 scopus 로고    scopus 로고
    • Neurite aggregation and calcium dysfunction in ipsc-derived sensory neurons with Parkinson’s disease-related LRRK2 G2019S mutation
    • Schwab AJ, Ebert AD. Neurite aggregation and calcium dysfunction in ipsc-derived sensory neurons with Parkinson’s disease-related LRRK2 G2019S mutation. Stem Cell Reports. 2015;5:1039–1052.
    • (2015) Stem Cell Reports , vol.5 , pp. 1039-1052
    • Schwab, A.J.1    Ebert, A.D.2
  • 51
    • 84996537122 scopus 로고    scopus 로고
    • Mutations in LRRK2 impair NF-kappaB pathway in iPSC-derived neurons
    • De Maturana L, Lang R,V, Zubiarrain A, et al. Mutations in LRRK2 impair NF-kappaB pathway in iPSC-derived neurons. J Neuroinflammation. 2016;13:295.
    • (2016) J Neuroinflammation , vol.13 , pp. 295
    • De Maturana, L.1    Lang, R.V.2    Zubiarrain, A.3
  • 52
    • 84941886639 scopus 로고    scopus 로고
    • I2020T mutant LRRK2 iPSC-derived neurons in the Sagamihara family exhibit increased Tau phosphorylation through the AKT/GSK-3beta signaling pathway
    • Ohta E, Nihira T, Uchino A, et al. I2020T mutant LRRK2 iPSC-derived neurons in the Sagamihara family exhibit increased Tau phosphorylation through the AKT/GSK-3beta signaling pathway. Hum Mol Genet. 2015;24:4879–4900.• An interesting report depicting in vitro results from patient-derived iPSC and its corresponding postmortem pathology from the same patient.
    • (2015) Hum Mol Genet , vol.24 , pp. 4879-4900
    • Ohta, E.1    Nihira, T.2    Uchino, A.3
  • 53
    • 84863584524 scopus 로고    scopus 로고
    • Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease
    • Cooper O, Seo H, Andrabi S, et al. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci Transl Med. 2012;4:141ra90.
    • (2012) Sci Transl Med , vol.4 , pp. 141ra90
    • Cooper, O.1    Seo, H.2    Andrabi, S.3
  • 54
    • 84869877601 scopus 로고    scopus 로고
    • Progressive degeneration of human neural stem cells caused by pathogenic LRRK2
    • Liu GH, Qu J, Suzuki K, et al. Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature. 2012;491:603–607.
    • (2012) Nature , vol.491 , pp. 603-607
    • Liu, G.H.1    Qu, J.2    Suzuki, K.3
  • 55
  • 56
    • 84865149238 scopus 로고    scopus 로고
    • GSK2578215A; a potent and highly selective 2-arylmethyloxy-5-substitutent-N-arylbenzamide LRRK2 kinase inhibitor
    • Reith AD, Bamborough P, Jandu K, et al. GSK2578215A; a potent and highly selective 2-arylmethyloxy-5-substitutent-N-arylbenzamide LRRK2 kinase inhibitor. Bioorg Med Chem Lett. 2012;22:5625–5629.
    • (2012) Bioorg Med Chem Lett , vol.22 , pp. 5625-5629
    • Reith, A.D.1    Bamborough, P.2    Jandu, K.3
  • 57
    • 84862777331 scopus 로고    scopus 로고
    • Characterization of TAE684 as a potent LRRK2 kinase inhibitor
    • Zhang J, Deng X, Choi HG, et al. Characterization of TAE684 as a potent LRRK2 kinase inhibitor. Bioorg Med Chem Lett. 2012;22:1864–1869.
    • (2012) Bioorg Med Chem Lett , vol.22 , pp. 1864-1869
    • Zhang, J.1    Deng, X.2    Choi, H.G.3
  • 58
    • 84929310825 scopus 로고    scopus 로고
    • Discovery of a pyrrolopyrimidine (JH-II-127), a highly potent, selective, and brain penetrant LRRK2 inhibitor
    • Hatcher JM, Zhang J, Choi HG, et al. Discovery of a pyrrolopyrimidine (JH-II-127), a highly potent, selective, and brain penetrant LRRK2 inhibitor. ACS Med Chem Lett. 2015;6:584–589.
    • (2015) ACS Med Chem Lett , vol.6 , pp. 584-589
    • Hatcher, J.M.1    Zhang, J.2    Choi, H.G.3
  • 59
    • 84870051317 scopus 로고    scopus 로고
    • Discovery of highly potent, selective, and brain-penetrable leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors
    • Estrada AA, Liu X, Baker-Glenn C, et al. Discovery of highly potent, selective, and brain-penetrable leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors. J Med Chem. 2012;55:9416–9433.
    • (2012) J Med Chem , vol.55 , pp. 9416-9433
    • Estrada, A.A.1    Liu, X.2    Baker-Glenn, C.3
  • 60
    • 84922674609 scopus 로고    scopus 로고
    • Effect of selective LRRK2 kinase inhibition on nonhuman primate lung
    • Fuji RN, Flagella M, Baca M, et al. Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci Transl Med. 2015;7:273ra15.•• This study highlights the importance of examining the potential safety risks of LRRK2 inhibitors in complex mammalian models compared to the commonly used murine models.
    • (2015) Sci Transl Med , vol.7 , pp. 273ra15
    • Fuji, R.N.1    Flagella, M.2    Baca, M.3
  • 61
    • 84894097684 scopus 로고    scopus 로고
    • Discovery of highly potent, selective, and brain-penetrant aminopyrazole leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors
    • Estrada AA, Chan BK, Baker-Glenn C, et al. Discovery of highly potent, selective, and brain-penetrant aminopyrazole leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors. J Med Chem. 2014;57:921–936.
    • (2014) J Med Chem , vol.57 , pp. 921-936
    • Estrada, A.A.1    Chan, B.K.2    Baker-Glenn, C.3
  • 62
    • 84920809166 scopus 로고    scopus 로고
    • 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
    • Henderson JL, Kormos BL, Hayward MM, 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;58:419–432.
    • (2015) J Med Chem , vol.58 , pp. 419-432
    • Henderson, J.L.1    Kormos, B.L.2    Hayward, M.M.3
  • 63
    • 84861595545 scopus 로고    scopus 로고
    • Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of alpha-synuclein and LRRK2 in the brain
    • Friedman LG, Lachenmayer ML, Wang J, et al. Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of alpha-synuclein and LRRK2 in the brain. J Neurosci. 2012;32:7585–7593.
    • (2012) J Neurosci , vol.32 , pp. 7585-7593
    • Friedman, L.G.1    Lachenmayer, M.L.2    Wang, J.3
  • 64
    • 77953090478 scopus 로고    scopus 로고
    • Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha-synuclein, and apoptotic cell death in aged mice
    • Tong Y, Yamaguchi H, Giaime E, 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 U S A. 2010;107:9879–9884.
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 9879-9884
    • Tong, Y.1    Yamaguchi, H.2    Giaime, E.3
  • 65
    • 84991112144 scopus 로고    scopus 로고
    • mTOR independent regulation of macroautophagy by leucine rich repeat kinase 2 via beclin-1
    • Manzoni C, Mamais A, Roosen DA, et al. mTOR independent regulation of macroautophagy by leucine rich repeat kinase 2 via beclin-1. Sci Rep. 2016;6:35106.
    • (2016) Sci Rep , vol.6 , pp. 35106
    • Manzoni, C.1    Mamais, A.2    Roosen, D.A.3
  • 66
    • 84992340883 scopus 로고    scopus 로고
    • Interplay between leucine-rich repeat kinase 2 (LRRK2) and p62/SQSTM-1 in selective autophagy
    • Park S, Han S, Choi I, et al. Interplay between leucine-rich repeat kinase 2 (LRRK2) and p62/SQSTM-1 in selective autophagy. Plos One. 2016;11:e0163029.
    • (2016) Plos One , vol.11 , pp. e0163029
    • Park, S.1    Han, S.2    Choi, I.3
  • 67
    • 84988569312 scopus 로고    scopus 로고
    • Pharmacological LRRK2 kinase inhibition induces LRRK2 protein destabilization and proteasomal degradation
    • Lobbestael E, Civiero L, De Wit T, et al. Pharmacological LRRK2 kinase inhibition induces LRRK2 protein destabilization and proteasomal degradation. Sci Rep. 2016;6:33897.
    • (2016) Sci Rep , vol.6 , pp. 33897
    • Lobbestael, E.1    Civiero, L.2    De Wit, T.3
  • 68
    • 84902163498 scopus 로고    scopus 로고
    • Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy
    • Schapansky J, Nardozzi JD, Felizia F, et al. Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy. Hum Mol Genet. 2014;23:4201–4214.
    • (2014) Hum Mol Genet , vol.23 , pp. 4201-4214
    • Schapansky, J.1    Nardozzi, J.D.2    Felizia, F.3
  • 69
    • 84873281274 scopus 로고    scopus 로고
    • RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson’s disease risk
    • MacLeod DA, Rhinn H, Kuwahara T, et al. RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson’s disease risk. Neuron. 2013;77:425–439.
    • (2013) Neuron , vol.77 , pp. 425-439
    • MacLeod, D.A.1    Rhinn, H.2    Kuwahara, T.3
  • 70
    • 84934967144 scopus 로고    scopus 로고
    • Alterations in late endocytic trafficking related to the pathobiology of LRRK2-linked Parkinson’s disease
    • Rivero-Rios P, Gomez-Suaga P, Fernandez B, et al. Alterations in late endocytic trafficking related to the pathobiology of LRRK2-linked Parkinson’s disease. Biochem Soc Trans. 2015;43:390–395.
    • (2015) Biochem Soc Trans , vol.43 , pp. 390-395
    • Rivero-Rios, P.1    Gomez-Suaga, P.2    Fernandez, B.3
  • 71
    • 30444436194 scopus 로고    scopus 로고
    • Vps9 domain-containing proteins: activators of Rab5 GTPases from yeast to neurons
    • Carney DS, Davies BA, Horazdovsky BF. Vps9 domain-containing proteins:activators of Rab5 GTPases from yeast to neurons. Trends Cell Biol. 2006;16:27–35.
    • (2006) Trends Cell Biol , vol.16 , pp. 27-35
    • Carney, D.S.1    Davies, B.A.2    Horazdovsky, B.F.3
  • 72
    • 0037598589 scopus 로고    scopus 로고
    • Role of Drosophila Rab5 during endosomal trafficking at the synapse and evoked neurotransmitter release
    • Wucherpfennig T, Wilsch-Brauninger M, Gonzalez-Gaitan M. Role of Drosophila Rab5 during endosomal trafficking at the synapse and evoked neurotransmitter release. J Cell Biol. 2003;161:609–624.
    • (2003) J Cell Biol , vol.161 , pp. 609-624
    • Wucherpfennig, T.1    Wilsch-Brauninger, M.2    Gonzalez-Gaitan, M.3
  • 73
    • 46549089664 scopus 로고    scopus 로고
    • LRRK2 regulates synaptic vesicle endocytosis
    • Shin N, Jeong H, Kwon J, et al. LRRK2 regulates synaptic vesicle endocytosis. Exp Cell Res. 2008;314:2055–2065.
    • (2008) Exp Cell Res , vol.314 , pp. 2055-2065
    • Shin, N.1    Jeong, H.2    Kwon, J.3
  • 74
    • 24144442691 scopus 로고    scopus 로고
    • Rab conversion as a mechanism of progression from early to late endosomes
    • Rink J, Ghigo E, Kalaidzidis Y, et al. Rab conversion as a mechanism of progression from early to late endosomes. Cell. 2005;122:735–749.
    • (2005) Cell , vol.122 , pp. 735-749
    • Rink, J.1    Ghigo, E.2    Kalaidzidis, Y.3
  • 75
    • 84863241584 scopus 로고    scopus 로고
    • Roles of the Drosophila LRRK2 homolog in Rab7-dependent lysosomal positioning
    • Dodson MW, Zhang T, Jiang C, et al. Roles of the Drosophila LRRK2 homolog in Rab7-dependent lysosomal positioning. Hum Mol Genet. 2012;21:1350–1363.
    • (2012) Hum Mol Genet , vol.21 , pp. 1350-1363
    • Dodson, M.W.1    Zhang, T.2    Jiang, C.3
  • 76
    • 84958567797 scopus 로고    scopus 로고
    • Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases
    • Steger M, Tonelli F, Ito G, et al. Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. Elife. 2016;5.
    • (2016) Elife , vol.5
    • Steger, M.1    Tonelli, F.2    Ito, G.3
  • 77
    • 70350653779 scopus 로고    scopus 로고
    • Substrate specificity and inhibitors of LRRK2, a protein kinase mutated in Parkinson’s disease
    • Nichols RJ, Dzamko N, Hutti JE, et al. Substrate specificity and inhibitors of LRRK2, a protein kinase mutated in Parkinson’s disease. Biochem J. 2009;424:47–60.
    • (2009) Biochem J , vol.424 , pp. 47-60
    • Nichols, R.J.1    Dzamko, N.2    Hutti, J.E.3
  • 78
    • 84865544952 scopus 로고    scopus 로고
    • Mitochondrial fission, fusion, and stress
    • Youle RJ, Van Der Bliek AM. Mitochondrial fission, fusion, and stress. Science. 2012;337:1062–1065.
    • (2012) Science , vol.337 , pp. 1062-1065
    • Youle, R.J.1    Van Der Bliek, A.M.2
  • 79
    • 84859259002 scopus 로고    scopus 로고
    • LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1
    • Wang X, Yan MH, Fujioka H, et al. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet. 2012;21:1931–1944.
    • (2012) Hum Mol Genet , vol.21 , pp. 1931-1944
    • Wang, X.1    Yan, M.H.2    Fujioka, H.3
  • 80
    • 84863728713 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 disturbs mitochondrial dynamics via dynamin-like protein
    • Niu J, Yu M, Wang C, et al. Leucine-rich repeat kinase 2 disturbs mitochondrial dynamics via dynamin-like protein. J Neurochem. 2012;122:650–658.
    • (2012) J Neurochem , vol.122 , pp. 650-658
    • Niu, J.1    Yu, M.2    Wang, C.3
  • 81
    • 84901017315 scopus 로고    scopus 로고
    • Mitochondrial dysfunction driven by the LRRK2-mediated pathway is associated with loss of Purkinje cells and motor coordination deficits in diabetic rat model
    • Yang S, Xia C, Li S, et al. Mitochondrial dysfunction driven by the LRRK2-mediated pathway is associated with loss of Purkinje cells and motor coordination deficits in diabetic rat model. Cell Death Dis. 2014;5:e1217.
    • (2014) Cell Death Dis , vol.5 , pp. e1217
    • Yang, S.1    Xia, C.2    Li, S.3
  • 82
    • 84897586124 scopus 로고    scopus 로고
    • Functional interaction of Parkinson’s disease-associated LRRK2 with members of the dynamin GTPase superfamily
    • Stafa K, Tsika E, Moser R, et al. Functional interaction of Parkinson’s disease-associated LRRK2 with members of the dynamin GTPase superfamily. Hum Mol Genet. 2014;23:2055–2077.
    • (2014) Hum Mol Genet , vol.23 , pp. 2055-2077
    • Stafa, K.1    Tsika, E.2    Moser, R.3
  • 83
    • 84994834600 scopus 로고    scopus 로고
    • Functional impairment in miro degradation and mitophagy is a shared feature in familial and sporadic Parkinson’s disease
    • Hsieh CH, Shaltouki A, Gonzalez AE, et al. Functional impairment in miro degradation and mitophagy is a shared feature in familial and sporadic Parkinson’s disease. Cell Stem Cell. 2016;19:709–724.
    • (2016) Cell Stem Cell , vol.19 , pp. 709-724
    • Hsieh, C.H.1    Shaltouki, A.2    Gonzalez, A.E.3
  • 84
    • 84941635707 scopus 로고    scopus 로고
    • LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase
    • Choi I, Kim B, Byun JW, et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun. 2015;6:8255.
    • (2015) Nat Commun , vol.6 , pp. 8255
    • Choi, I.1    Kim, B.2    Byun, J.W.3
  • 85
    • 84920426451 scopus 로고    scopus 로고
    • Increasing microtubule acetylation rescues axonal transport and locomotor deficits caused by LRRK2 Roc-COR domain mutations
    • Godena VK, Brookes-Hocking N, Moller A, et al. Increasing microtubule acetylation rescues axonal transport and locomotor deficits caused by LRRK2 Roc-COR domain mutations. Nat Commun. 2014;5:5245.
    • (2014) Nat Commun , vol.5 , pp. 5245
    • Godena, V.K.1    Brookes-Hocking, N.2    Moller, A.3
  • 86
    • 84955212896 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 interacts with p21-activated kinase 6 to control neurite complexity in mammalian brain
    • Civiero L, Cirnaru MD, Beilina A, et al. Leucine-rich repeat kinase 2 interacts with p21-activated kinase 6 to control neurite complexity in mammalian brain. J Neurochem. 2015;135:1242–1256.
    • (2015) J Neurochem , vol.135 , pp. 1242-1256
    • Civiero, L.1    Cirnaru, M.D.2    Beilina, A.3
  • 87
    • 84856404449 scopus 로고    scopus 로고
    • LRRK2 phosphorylates tubulin-associated tau but not the free molecule: LRRK2-mediated regulation of the tau-tubulin association and neurite outgrowth
    • Kawakami F, Yabata T, Ohta E, et al. LRRK2 phosphorylates tubulin-associated tau but not the free molecule:LRRK2-mediated regulation of the tau-tubulin association and neurite outgrowth. Plos One. 2012;7:e30834.
    • (2012) Plos One , vol.7 , pp. e30834
    • Kawakami, F.1    Yabata, T.2    Ohta, E.3
  • 88
    • 84939467302 scopus 로고    scopus 로고
    • Physiologically relevant factors influence tau phosphorylation by leucine-rich repeat kinase 2
    • Hamm M, Bailey R, Shaw G, et al. Physiologically relevant factors influence tau phosphorylation by leucine-rich repeat kinase 2. J Neurosci Res. 2015;93:1567–1580.
    • (2015) J Neurosci Res , vol.93 , pp. 1567-1580
    • Hamm, M.1    Bailey, R.2    Shaw, G.3
  • 89
    • 84939609978 scopus 로고    scopus 로고
    • LRRK2 facilitates tau phosphorylation through strong interaction with tau and cdk5
    • Shanley MR, Hawley D, Leung S, et al. LRRK2 facilitates tau phosphorylation through strong interaction with tau and cdk5. Biochemistry. 2015;54:5198–5208.
    • (2015) Biochemistry , vol.54 , pp. 5198-5208
    • Shanley, M.R.1    Hawley, D.2    Leung, S.3
  • 90
    • 84930155887 scopus 로고    scopus 로고
    • LRRK2 promotes tau accumulation, aggregation and release
    • Guerreiro PS, Gerhardt E, Lopes Da Fonseca T, et al. LRRK2 promotes tau accumulation, aggregation and release. Mol Neurobiol. 2016;53:3124–3135.
    • (2016) Mol Neurobiol , vol.53 , pp. 3124-3135
    • Guerreiro, P.S.1    Gerhardt, E.2    Lopes Da Fonseca, T.3
  • 91
    • 84940421908 scopus 로고    scopus 로고
    • Chemical genetic approach identifies microtubule affinity-regulating kinase 1 as a leucine-rich repeat kinase 2 substrate
    • Krumova P, Reyniers L, Meyer M, et al. Chemical genetic approach identifies microtubule affinity-regulating kinase 1 as a leucine-rich repeat kinase 2 substrate. Faseb J. 2015;29:2980–2992.
    • (2015) Faseb J , vol.29 , pp. 2980-2992
    • Krumova, P.1    Reyniers, L.2    Meyer, M.3
  • 92
    • 84891923527 scopus 로고    scopus 로고
    • A direct interaction between leucine-rich repeat kinase 2 and specific beta-tubulin isoforms regulates tubulin acetylation
    • Law BM, Spain VA, Leinster VH, et al. A direct interaction between leucine-rich repeat kinase 2 and specific beta-tubulin isoforms regulates tubulin acetylation. J Biol Chem. 2014;289:895–908.
    • (2014) J Biol Chem , vol.289 , pp. 895-908
    • Law, B.M.1    Spain, V.A.2    Leinster, V.H.3
  • 93
    • 84949650757 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 positively regulates inflammation and down-regulates NF-kappaB p50 signaling in cultured microglia cells
    • Russo I, Berti G, Plotegher N, et al. Leucine-rich repeat kinase 2 positively regulates inflammation and down-regulates NF-kappaB p50 signaling in cultured microglia cells. J Neuroinflammation. 2015;12:230.
    • (2015) J Neuroinflammation , vol.12 , pp. 230
    • Russo, I.1    Berti, G.2    Plotegher, N.3
  • 94
    • 78149473340 scopus 로고    scopus 로고
    • LRRK2 is involved in the IFN-gamma response and host response to pathogens
    • Gardet A, Benita Y, Li C, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185:5577–5585.
    • (2010) J Immunol , vol.185 , pp. 5577-5585
    • Gardet, A.1    Benita, Y.2    Li, C.3
  • 95
    • 84902360686 scopus 로고    scopus 로고
    • Interferon-gamma induces leucine-rich repeat kinase LRRK2 via extracellular signal-regulated kinase ERK5 in macrophages
    • Kuss M, Adamopoulou E, Kahle PJ. Interferon-gamma induces leucine-rich repeat kinase LRRK2 via extracellular signal-regulated kinase ERK5 in macrophages. J Neurochem. 2014;129:980–987.
    • (2014) J Neurochem , vol.129 , pp. 980-987
    • Kuss, M.1    Adamopoulou, E.2    Kahle, P.J.3
  • 96
    • 85018214550 scopus 로고    scopus 로고
    • The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins
    • Moehle MS, Daher JP, Hull TD, et al. The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins. Hum Mol Genet. 2015;24:4250–4267.
    • (2015) Hum Mol Genet , vol.24 , pp. 4250-4267
    • Moehle, M.S.1    Daher, J.P.2    Hull, T.D.3
  • 97
    • 84862497413 scopus 로고    scopus 로고
    • The kappaB kinase family phosphorylates the Parkinson’s disease kinase LRRK2 at Ser935 and Ser910 during toll-like receptor signaling
    • Dzamko N, Inesta-Vaquera F, Zhang J, et al. The kappaB kinase family phosphorylates the Parkinson’s disease kinase LRRK2 at Ser935 and Ser910 during toll-like receptor signaling. Plos One. 2012;7:e39132.
    • (2012) Plos One , vol.7 , pp. e39132
    • Dzamko, N.1    Inesta-Vaquera, F.2    Zhang, J.3
  • 98
    • 84858620880 scopus 로고    scopus 로고
    • Parkinson’s disease-linked leucine-rich repeat kinase 2(R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity
    • 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.
    • (2012) Neuroscience , vol.208 , pp. 41-48
    • Gillardon, F.1    Schmid, R.2    Draheim, H.3
  • 99
    • 84856632181 scopus 로고    scopus 로고
    • LRRK2 inhibition attenuates microglial inflammatory responses
    • Moehle MS, Webber PJ, Tse T, et al. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32:1602–1611.
    • (2012) J Neurosci , vol.32 , pp. 1602-1611
    • Moehle, M.S.1    Webber, P.J.2    Tse, T.3
  • 100
    • 84969921143 scopus 로고    scopus 로고
    • Inflammatory profile in LRRK2-associated prodromal and clinical PD
    • Brockmann K, Apel A, Schulte C, et al. Inflammatory profile in LRRK2-associated prodromal and clinical PD. J Neuroinflammation. 2016;13:122.• An interesting report comparing prodromal and clinical PD, which also shows the widening scope of current LRRK2 research.
    • (2016) J Neuroinflammation , vol.13 , pp. 122
    • Brockmann, K.1    Apel, A.2    Schulte, C.3
  • 101
    • 80054977424 scopus 로고    scopus 로고
    • Chemoproteomics-based design of potent LRRK2-selective lead compounds that attenuate Parkinson’s disease-related toxicity in human neurons
    • Ramsden N, Perrin J, Ren Z, 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;6:1021–1028.
    • (2011) ACS Chem Biol , vol.6 , pp. 1021-1028
    • Ramsden, N.1    Perrin, J.2    Ren, Z.3
  • 102
    • 84893877488 scopus 로고    scopus 로고
    • Phosphoproteomic evaluation of pharmacological inhibition of leucine-rich repeat kinase 2 reveals significant off-target effects of LRRK-2-IN-1
    • Luerman GC, Nguyen C, Samaroo H, et al. Phosphoproteomic evaluation of pharmacological inhibition of leucine-rich repeat kinase 2 reveals significant off-target effects of LRRK-2-IN-1. J Neurochem. 2014;128:561–576.•• Inhibitors are widely used in current research; this increases awareness about drugs’ off-target effects. This is highlighted as LRRK2-IN-1 was a commonly employed LRRK2 inhibitor until its off-target effects were reported.
    • (2014) J Neurochem , vol.128 , pp. 561-576
    • Luerman, G.C.1    Nguyen, C.2    Samaroo, H.3
  • 103
    • 33846562487 scopus 로고    scopus 로고
    • Parkinson’s disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity
    • West AB, Moore DJ, Choi C, et al. Parkinson’s disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity. Hum Mol Genet. 2007;16:223–232.
    • (2007) Hum Mol Genet , vol.16 , pp. 223-232
    • West, A.B.1    Moore, D.J.2    Choi, C.3
  • 104
    • 20444489689 scopus 로고    scopus 로고
    • Motor fluctuations and dyskinesias in Parkinson’s disease: clinical manifestations
    • Jankovic J. Motor fluctuations and dyskinesias in Parkinson’s disease:clinical manifestations. Mov Disord. 2005;20(Suppl 11):S11–6.
    • (2005) Mov Disord , vol.20 , pp. S11-S16
    • Jankovic, J.1
  • 105
    • 34248222587 scopus 로고    scopus 로고
    • Levodopa, motor fluctuations and dyskinesia in Parkinson’s disease
    • Muller T, Russ H. Levodopa, motor fluctuations and dyskinesia in Parkinson’s disease. Expert Opin Pharmacother. 2006;7:1715–1730.
    • (2006) Expert Opin Pharmacother , vol.7 , pp. 1715-1730
    • Muller, T.1    Russ, H.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.