Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2018

Open Access 01-12-2018 | Research article

Parkinson disease-associated mutations in LRRK2 cause centrosomal defects via Rab8a phosphorylation

Authors: Jesús Madero-Pérez, Elena Fdez, Belén Fernández, Antonio J. Lara Ordóñez, Marian Blanca Ramírez, Patricia Gómez-Suaga, Dieter Waschbüsch, Evy Lobbestael, Veerle Baekelandt, Angus C. Nairn, Javier Ruiz-Martínez, Ana Aiastui, Adolfo López de Munain, Pawel Lis, Thomas Comptdaer, Jean-Marc Taymans, Marie-Christine Chartier-Harlin, Alexandria Beilina, Adriano Gonnelli, Mark R. Cookson, Elisa Greggio, Sabine Hilfiker

Published in: Molecular Neurodegeneration | Issue 1/2018

Login to get access

Abstract

Background

Mutations in LRRK2 are a common genetic cause of Parkinson’s disease (PD). LRRK2 interacts with and phosphorylates a subset of Rab proteins including Rab8a, a protein which has been implicated in various centrosome-related events. However, the cellular consequences of such phosphorylation remain elusive.

Methods

Human neuroblastoma SH-SY5Y cells stably expressing wildtype or pathogenic LRRK2 were used to test for polarity defects in the context of centrosomal positioning. Centrosomal cohesion deficits were analyzed from transiently transfected HEK293T cells, as well as from two distinct peripheral cell types derived from LRRK2-PD patients. Kinase assays, coimmunoprecipitation and GTP binding/retention assays were used to address Rab8a phosphorylation by LRRK2 and its effects in vitro. Transient transfections and siRNA experiments were performed to probe for the implication of Rab8a and its phosphorylated form in the centrosomal deficits caused by pathogenic LRRK2.

Results

Here, we show that pathogenic LRRK2 causes deficits in centrosomal positioning with effects on neurite outgrowth, cell polarization and directed migration. Pathogenic LRRK2 also causes deficits in centrosome cohesion which can be detected in peripheral cells derived from LRRK2-PD patients as compared to healthy controls, and which are reversed upon LRRK2 kinase inhibition. The centrosomal cohesion and polarity deficits can be mimicked when co-expressing wildtype LRRK2 with wildtype but not phospho-deficient Rab8a. The centrosomal defects induced by pathogenic LRRK2 are associated with a kinase activity-dependent increase in the centrosomal localization of phosphorylated Rab8a, and are prominently reduced upon RNAi of Rab8a.

Conclusions

Our findings reveal a new function of LRRK2 mediated by Rab8a phosphorylation and related to various centrosomal defects.
Appendix
Available only for authorised users
Literature
3.
go back to reference Steger M, Tonelli F, Ito G, Davies P, Trost M, Vetter M, Wachter S, Lorentzen E, Duddy G, Wilson S, et al. Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of Rab GTPases. eLife. 2016;5:e12813. Steger M, Tonelli F, Ito G, Davies P, Trost M, Vetter M, Wachter S, Lorentzen E, Duddy G, Wilson S, et al. Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of Rab GTPases. eLife. 2016;5:e12813.
4.
go back to reference MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93.CrossRefPubMed MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93.CrossRefPubMed
5.
go back to reference Gomez-Suaga P, Luzon-Toro B, Churamani D, Zhang L, Bloor-Young D, Patel S, Woodman PG, Churchill GC, Hilfiker S. Leucine-rich repeat kinase 2 regulates autophagy through a calcium-dependent pathway involving NAADP. Hum Mol Genet. 2012;21:511–25.CrossRefPubMed Gomez-Suaga P, Luzon-Toro B, Churamani D, Zhang L, Bloor-Young D, Patel S, Woodman PG, Churchill GC, Hilfiker S. Leucine-rich repeat kinase 2 regulates autophagy through a calcium-dependent pathway involving NAADP. Hum Mol Genet. 2012;21:511–25.CrossRefPubMed
6.
go back to reference Sanchez-Danes A, Richaud-Patin Y, Carballo-Carbajal I, Jimenez-Delgado S, Caig C, Mora S, Di Guglielmo C, Ezquerra M, Patel B, Giralt A, et al. Disease-specific phenotypes in dopamine neurons from human iPS-based models of genetic and sporadic Parkinson's disease. EMBO Mol Med. 2012;4:380–95.CrossRefPubMedPubMedCentral Sanchez-Danes A, Richaud-Patin Y, Carballo-Carbajal I, Jimenez-Delgado S, Caig C, Mora S, Di Guglielmo C, Ezquerra M, Patel B, Giralt A, et al. Disease-specific phenotypes in dopamine neurons from human iPS-based models of genetic and sporadic Parkinson's disease. EMBO Mol Med. 2012;4:380–95.CrossRefPubMedPubMedCentral
7.
go back to reference MacLeod DA, Rhinn H, Kuwahara T, Zolin A, Di Paolo G, McCabe BD, Marder KS, Honig LS, Clark LN, Small SA, Abeliovich A. RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson's disease risk. Neuron. 2013;77:425–39.CrossRefPubMedPubMedCentral MacLeod DA, Rhinn H, Kuwahara T, Zolin A, Di Paolo G, McCabe BD, Marder KS, Honig LS, Clark LN, Small SA, Abeliovich A. RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson's disease risk. Neuron. 2013;77:425–39.CrossRefPubMedPubMedCentral
8.
go back to reference Gomez-Suaga P, Rivero-Rios P, Fdez E, Blanca Ramirez M, Ferrer I, Aiastui A, Lopez De Munain A, Hilfiker S. LRRK2 delays degradative receptor trafficking by impeding late endosomal budding through decreasing Rab7 activity. Hum Mol Genet. 2014;23:6779–96.CrossRefPubMed Gomez-Suaga P, Rivero-Rios P, Fdez E, Blanca Ramirez M, Ferrer I, Aiastui A, Lopez De Munain A, Hilfiker S. LRRK2 delays degradative receptor trafficking by impeding late endosomal budding through decreasing Rab7 activity. Hum Mol Genet. 2014;23:6779–96.CrossRefPubMed
9.
go back to reference Beilina A, Rudenko IN, Kaganovich A, Civiero L, Chau H, Kalia SK, Kalia LV, Lobbestael E, Chia R, Ndukwe K, 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 U S A. 2014;111:2626–31.CrossRefPubMedPubMedCentral Beilina A, Rudenko IN, Kaganovich A, Civiero L, Chau H, Kalia SK, Kalia LV, Lobbestael E, Chia R, Ndukwe K, 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 U S A. 2014;111:2626–31.CrossRefPubMedPubMedCentral
10.
go back to reference Parisiadou L, Xie C, Cho HJ, Lin X, Gu XL, Long CX, Lobbestael E, Baekelandt V, Taymans JM, Sun L, Cai H. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci. 2009;29:13971–80.CrossRefPubMedPubMedCentral Parisiadou L, Xie C, Cho HJ, Lin X, Gu XL, Long CX, Lobbestael E, Baekelandt V, Taymans JM, Sun L, Cai H. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci. 2009;29:13971–80.CrossRefPubMedPubMedCentral
12.
go back to reference Caesar M, Zach S, Carlson CB, Brockmann K, Gasser T, Gillardon F. Leucine-rich repeat kinase 2 functionally interacts with microtubules and kinase-dependently modulates cell migration. Neurobiol Dis. 2013;54:280–8.CrossRefPubMed Caesar M, Zach S, Carlson CB, Brockmann K, Gasser T, Gillardon F. Leucine-rich repeat kinase 2 functionally interacts with microtubules and kinase-dependently modulates cell migration. Neurobiol Dis. 2013;54:280–8.CrossRefPubMed
13.
go back to reference Moehle MS, Daher JP, Hull TD, Boddu R, Abdelmotilib HA, Mobley J, Kannarkat GT, Tansey MG, West AB. The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins. Hum Mol Genet. 2015;24:4250–67.CrossRefPubMedPubMedCentral Moehle MS, Daher JP, Hull TD, Boddu R, Abdelmotilib HA, Mobley J, Kannarkat GT, Tansey MG, West AB. The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins. Hum Mol Genet. 2015;24:4250–67.CrossRefPubMedPubMedCentral
14.
go back to reference Choi I, Kim B, Byun JW, Baik SH, Huh YH, Kim JH, Mook-Jung I, Song WK, Shin JH, Seo H, et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun. 2015;6:8255.CrossRefPubMedPubMedCentral Choi I, Kim B, Byun JW, Baik SH, Huh YH, Kim JH, Mook-Jung I, Song WK, Shin JH, Seo H, et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun. 2015;6:8255.CrossRefPubMedPubMedCentral
15.
go back to reference Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, Braithwaite AT, Ogholikhan S, Aigner R, Winkler J, et al. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011;41:706–16.CrossRefPubMed Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, Braithwaite AT, Ogholikhan S, Aigner R, Winkler J, et al. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011;41:706–16.CrossRefPubMed
16.
go back to reference Liu GH, Qu J, Suzuki K, Nivet E, Li M, Montserrat N, Yi F, Xu X, Ruiz S, Zhang W, et al. Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature. 2012;491:603–7.CrossRefPubMedPubMedCentral Liu GH, Qu J, Suzuki K, Nivet E, Li M, Montserrat N, Yi F, Xu X, Ruiz S, Zhang W, et al. Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature. 2012;491:603–7.CrossRefPubMedPubMedCentral
17.
go back to reference Thaler A, Mirelman A, Gurevich T, Simon E, Orr-Urtreger A, Marder K, Bressman S, Giladi N, Consortium LAJ. Lower cognitive performance in healthy G2019S LRRK2 mutation carriers. Neurology. 2012;79:1027–32.CrossRefPubMedPubMedCentral Thaler A, Mirelman A, Gurevich T, Simon E, Orr-Urtreger A, Marder K, Bressman S, Giladi N, Consortium LAJ. Lower cognitive performance in healthy G2019S LRRK2 mutation carriers. Neurology. 2012;79:1027–32.CrossRefPubMedPubMedCentral
18.
go back to reference Mirelman A, Alcalay RN, Saunders-Pullman R, Yasinovsky K, Thaler A, Gurevich T, Mejia-Santana H, Raymond D, Gana-Weisz M, Bar-Shira A, et al. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov Disord. 2015;30:981–6.CrossRefPubMedPubMedCentral Mirelman A, Alcalay RN, Saunders-Pullman R, Yasinovsky K, Thaler A, Gurevich T, Mejia-Santana H, Raymond D, Gana-Weisz M, Bar-Shira A, et al. Nonmotor symptoms in healthy Ashkenazi Jewish carriers of the G2019S mutation in the LRRK2 gene. Mov Disord. 2015;30:981–6.CrossRefPubMedPubMedCentral
19.
go back to reference Alegre-Abarrategui J, Christian H, Lufino MM, Mutihac R, Venda LL, Ansorge O, Wade-Martins R. LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum Mol Genet. 2009;18:4022–34.CrossRefPubMedPubMedCentral Alegre-Abarrategui J, Christian H, Lufino MM, Mutihac R, Venda LL, Ansorge O, Wade-Martins R. LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum Mol Genet. 2009;18:4022–34.CrossRefPubMedPubMedCentral
20.
go back to reference Hehnly H, Chen CT, Powers CM, Liu HL, Doxsey S. The centrosome regulates the Rab11- dependent recycling endosome pathway at appendages of the mother centriole. Curr Biol. 2012;22:1944–50.CrossRefPubMedPubMedCentral Hehnly H, Chen CT, Powers CM, Liu HL, Doxsey S. The centrosome regulates the Rab11- dependent recycling endosome pathway at appendages of the mother centriole. Curr Biol. 2012;22:1944–50.CrossRefPubMedPubMedCentral
21.
go back to reference Jakobsen L, Vanselow K, Skogs M, Toyoda Y, Lundberg E, Poser I, Falkenby LG, Bennetzen M, Westendorf J, Nigg EA, et al. Novel asymmetrically localizing components of human centrosomes identified by complementary proteomics methods. EMBO J. 2011;30:1520–35.CrossRefPubMedPubMedCentral Jakobsen L, Vanselow K, Skogs M, Toyoda Y, Lundberg E, Poser I, Falkenby LG, Bennetzen M, Westendorf J, Nigg EA, et al. Novel asymmetrically localizing components of human centrosomes identified by complementary proteomics methods. EMBO J. 2011;30:1520–35.CrossRefPubMedPubMedCentral
22.
go back to reference Westlake CJ, Baye LM, Nachury MV, Wright KJ, Ervin KE, Phu L, Chalouni C, Beck JS, Kirkpatrick DS, Slusarski DC, et al. Primary cilia membrane assembly is initiated by Rab11 and transport protein particle II (TRAPPII) complex-dependent trafficking of Rabin8 to the centrosome. Proc Natl Acad Sci U S A. 2011;108:2759–64.CrossRefPubMedPubMedCentral Westlake CJ, Baye LM, Nachury MV, Wright KJ, Ervin KE, Phu L, Chalouni C, Beck JS, Kirkpatrick DS, Slusarski DC, et al. Primary cilia membrane assembly is initiated by Rab11 and transport protein particle II (TRAPPII) complex-dependent trafficking of Rabin8 to the centrosome. Proc Natl Acad Sci U S A. 2011;108:2759–64.CrossRefPubMedPubMedCentral
23.
go back to reference de Anda FC, Pollarolo G, Da Silva JS, Camoletto PG, Feiguin F, Dotti CG. Centrosome localization determines neuronal polarity. Nature. 2005;436:704–8.CrossRefPubMed de Anda FC, Pollarolo G, Da Silva JS, Camoletto PG, Feiguin F, Dotti CG. Centrosome localization determines neuronal polarity. Nature. 2005;436:704–8.CrossRefPubMed
25.
go back to reference Elric J, Etienne-Manneville S. Centrosome positioning in polarized cells: common themes and variations. Exp Cell Res. 2014;328:240–8.CrossRefPubMed Elric J, Etienne-Manneville S. Centrosome positioning in polarized cells: common themes and variations. Exp Cell Res. 2014;328:240–8.CrossRefPubMed
28.
go back to reference Reyniers L, Del Giudice MG, Civiero L, Belluzzi E, Lobbestael E, Beilina A, Arrigoni G, Derua R, Waelkens E, Li Y, et al. Differential protein-protein interactions of LRRK1 and LRRK2 indicate roles in distinct cellular signaling pathways. J Neurochem. 2014;131:239–50.CrossRefPubMedPubMedCentral Reyniers L, Del Giudice MG, Civiero L, Belluzzi E, Lobbestael E, Beilina A, Arrigoni G, Derua R, Waelkens E, Li Y, et al. Differential protein-protein interactions of LRRK1 and LRRK2 indicate roles in distinct cellular signaling pathways. J Neurochem. 2014;131:239–50.CrossRefPubMedPubMedCentral
29.
go back to reference Vancraenenbroeck R, De Raeymaecker J, Lobbestael E, Gao F, De Maeyer M, Voet A, Baekelandt V, Taymans JM. In silico, in vitro and cellular analysis with a kinome-wide inhibitor panel correlates cellular LRRK2 dephosphorylation to inhibitor activity on LRRK2. Front Mol Neurosci. 2014;7(51) Vancraenenbroeck R, De Raeymaecker J, Lobbestael E, Gao F, De Maeyer M, Voet A, Baekelandt V, Taymans JM. In silico, in vitro and cellular analysis with a kinome-wide inhibitor panel correlates cellular LRRK2 dephosphorylation to inhibitor activity on LRRK2. Front Mol Neurosci. 2014;7(51)
30.
go back to reference Louie LG, King MC. A novel approach to establishing permanent lymphoblastoid cell lines: Epstein-Barr virus transformation of cryopreserved lymphocytes. Am J Hum Genet. 1991;48:637–8.PubMedPubMedCentral Louie LG, King MC. A novel approach to establishing permanent lymphoblastoid cell lines: Epstein-Barr virus transformation of cryopreserved lymphocytes. Am J Hum Genet. 1991;48:637–8.PubMedPubMedCentral
31.
go back to reference Lindqvist A, van Zon W, Karlsson Rosenthal C, Wolthuis RM. Cyclin B1-Cdk1 activation continues after centrosome separation to control mitotic progression. PLoS Biol. 2007;5:e123.CrossRefPubMedPubMedCentral Lindqvist A, van Zon W, Karlsson Rosenthal C, Wolthuis RM. Cyclin B1-Cdk1 activation continues after centrosome separation to control mitotic progression. PLoS Biol. 2007;5:e123.CrossRefPubMedPubMedCentral
32.
go back to reference Abbi S, Ueda H, Zheng C, Cooper LA, Zhao J, Christopher R, Guan JL. Regulation of focal adhesion kinase by a novel protein inhibitor FIP200. Mol Biol Cell. 2002;13:3178–91.CrossRefPubMedPubMedCentral Abbi S, Ueda H, Zheng C, Cooper LA, Zhao J, Christopher R, Guan JL. Regulation of focal adhesion kinase by a novel protein inhibitor FIP200. Mol Biol Cell. 2002;13:3178–91.CrossRefPubMedPubMedCentral
33.
go back to reference Etienne-Manneville S, Hall A. Integrin-mediated activation of Cdc42 controls cell polarity in migrating astrocytes through PKCzeta. Cell. 2001;106:489–98.CrossRefPubMed Etienne-Manneville S, Hall A. Integrin-mediated activation of Cdc42 controls cell polarity in migrating astrocytes through PKCzeta. Cell. 2001;106:489–98.CrossRefPubMed
34.
go back to reference Taymans JM, Vancraenenbroeck R, Ollikainen P, Beilina A, Lobbestael E, De Maeyer M, Baekelandt V, Cookson MR. LRRK2 kinase activity is dependent on LRRK2 GTP binding capacity but independent of LRRK2 GTP binding. PLoS One. 2011;6:e23207.CrossRefPubMedPubMedCentral Taymans JM, Vancraenenbroeck R, Ollikainen P, Beilina A, Lobbestael E, De Maeyer M, Baekelandt V, Cookson MR. LRRK2 kinase activity is dependent on LRRK2 GTP binding capacity but independent of LRRK2 GTP binding. PLoS One. 2011;6:e23207.CrossRefPubMedPubMedCentral
36.
go back to reference Hurtado L, Caballero C, Gavilan MP, Cardenas J, Bornens M, Rios RM. Disconnecting the Golgi ribbon from the centrosome prevents directional cell migration and ciliogenesis. J Cell Biol. 2011;193:917–33.CrossRefPubMedPubMedCentral Hurtado L, Caballero C, Gavilan MP, Cardenas J, Bornens M, Rios RM. Disconnecting the Golgi ribbon from the centrosome prevents directional cell migration and ciliogenesis. J Cell Biol. 2011;193:917–33.CrossRefPubMedPubMedCentral
37.
go back to reference Deng X, Dzamko N, Prescott A, Davies P, Liu Q, Yang Q, Lee JD, Patricelli MP, Nomanbhoy TK, Alessi DR, Gray NS. Characterization of a selective inhibitor of the Parkinson's disease kinase LRRK2. Nat Chem Biol. 2011;7:203–5.CrossRefPubMedPubMedCentral Deng X, Dzamko N, Prescott A, Davies P, Liu Q, Yang Q, Lee JD, Patricelli MP, Nomanbhoy TK, Alessi DR, Gray NS. Characterization of a selective inhibitor of the Parkinson's disease kinase LRRK2. Nat Chem Biol. 2011;7:203–5.CrossRefPubMedPubMedCentral
38.
go back to reference Reith AD, Bamborough P, Jandu K, Andreotti D, Mensah L, Dossang P, Choi HG, Deng X, Zhang J, Alessi DR, Gray NS. GSK2578215A; a potent and highly selective 2-arylmethyloxy-5-substitutent-N-arylbenzamide LRRK2 kinase inhibitor. Bioorg Med Chem Lett. 2012;22:5625–9.CrossRefPubMedPubMedCentral Reith AD, Bamborough P, Jandu K, Andreotti D, Mensah L, Dossang P, Choi HG, Deng X, Zhang J, Alessi DR, Gray NS. GSK2578215A; a potent and highly selective 2-arylmethyloxy-5-substitutent-N-arylbenzamide LRRK2 kinase inhibitor. Bioorg Med Chem Lett. 2012;22:5625–9.CrossRefPubMedPubMedCentral
39.
go back to reference Kett LR, Boassa D, Ho CC, Rideout HJ, Hu J, Terada M, Ellisman M, Dauer WT. LRRK2 Parkinson disease mutations enhance its microtubule association. Hum Mol Genet. 2012;21:890–9.CrossRefPubMed Kett LR, Boassa D, Ho CC, Rideout HJ, Hu J, Terada M, Ellisman M, Dauer WT. LRRK2 Parkinson disease mutations enhance its microtubule association. Hum Mol Genet. 2012;21:890–9.CrossRefPubMed
40.
go back to reference Papkovskaia TD, Chau KY, Inesta-Vaquera F, Papkovsky DB, Healy DG, Nishio K, Staddon J, Duchen MR, Hardy J, Schapira AH, Cooper JM. G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization. Hum Mol Genet. 2012;21:4201–13.CrossRefPubMedPubMedCentral Papkovskaia TD, Chau KY, Inesta-Vaquera F, Papkovsky DB, Healy DG, Nishio K, Staddon J, Duchen MR, Hardy J, Schapira AH, Cooper JM. G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization. Hum Mol Genet. 2012;21:4201–13.CrossRefPubMedPubMedCentral
41.
go back to reference Fell MJ, Mirescu C, Basu K, Cheewatrakoolpong B, DeMong DE, Ellis JM, Hyde LA, Lin Y, Markgraf CG, Mei H, 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.CrossRefPubMed Fell MJ, Mirescu C, Basu K, Cheewatrakoolpong B, DeMong DE, Ellis JM, Hyde LA, Lin Y, Markgraf CG, Mei H, 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.CrossRefPubMed
42.
go back to reference Shisheva A, Chinni SR, DeMarco C. General role of GDP dissociation inhibitor 2 in membrane release of Rab proteins: modulations of its functional interactions by in vitro and in vivo structural modifications. Biochemistry. 1999;38:11711–21.CrossRefPubMed Shisheva A, Chinni SR, DeMarco C. General role of GDP dissociation inhibitor 2 in membrane release of Rab proteins: modulations of its functional interactions by in vitro and in vivo structural modifications. Biochemistry. 1999;38:11711–21.CrossRefPubMed
45.
go back to reference Rios RM. The centrosome-Golgi apparatus nexus. Phil Trans R Soc B. 2014;369:20130462. Rios RM. The centrosome-Golgi apparatus nexus. Phil Trans R Soc B. 2014;369:20130462.
46.
47.
go back to reference Nichols RJ, Dzamko N, Morrice NA, Campbell DG, Deak M, Ordureau A, Macartney T, Tong Y, Shen J, Prescott AR, Alessi DR. 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.CrossRefPubMedPubMedCentral Nichols RJ, Dzamko N, Morrice NA, Campbell DG, Deak M, Ordureau A, Macartney T, Tong Y, Shen J, Prescott AR, Alessi DR. 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.CrossRefPubMedPubMedCentral
48.
go back to reference Dzamko N, Deak M, Hentati F, Reith AD, Prescott AR, Alessi DR, Nichols RJ. 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;430:405–13.CrossRefPubMedPubMedCentral Dzamko N, Deak M, Hentati F, Reith AD, Prescott AR, Alessi DR, Nichols RJ. 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;430:405–13.CrossRefPubMedPubMedCentral
49.
go back to reference Li X, Wang QJ, Pan N, Lee S, Zhao Y, Chait BT, Yue Z. Phosphorylation-dependent 14-3-3 binding to LRRK2 is impaired by common mutations of familial Parkinson's disease. PLoS One. 2011;6:e17153.CrossRefPubMedPubMedCentral Li X, Wang QJ, Pan N, Lee S, Zhao Y, Chait BT, Yue Z. Phosphorylation-dependent 14-3-3 binding to LRRK2 is impaired by common mutations of familial Parkinson's disease. PLoS One. 2011;6:e17153.CrossRefPubMedPubMedCentral
50.
go back to reference 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;120:37–45.CrossRefPubMed 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;120:37–45.CrossRefPubMed
51.
go back to reference Lobbestael E, Zhao J, Rudenko IN, Beylina A, Gao F, Wetter J, Beullens M, Bollen M, Cookson MR, Baekelandt V, et al. Identification of protein phosphatase 1 as a regulator of the LRRK2 phosphorylation cycle. Biochem J. 2013;456:119–28.CrossRefPubMedPubMedCentral Lobbestael E, Zhao J, Rudenko IN, Beylina A, Gao F, Wetter J, Beullens M, Bollen M, Cookson MR, Baekelandt V, et al. Identification of protein phosphatase 1 as a regulator of the LRRK2 phosphorylation cycle. Biochem J. 2013;456:119–28.CrossRefPubMedPubMedCentral
52.
go back to reference Reynolds A, Doggett EA, Riddle SM, Lebakken CS, Nichols RJ. LRRK2 kinase activity and biology are not uniformly predicted by its autophosphorylation and cellular phosphorylation site status. Front Mol Neurosci. 2014;7(54) Reynolds A, Doggett EA, Riddle SM, Lebakken CS, Nichols RJ. LRRK2 kinase activity and biology are not uniformly predicted by its autophosphorylation and cellular phosphorylation site status. Front Mol Neurosci. 2014;7(54)
53.
go back to reference Blanca Ramirez M, Lara Ordonez AJ, Fdez E, Madero-Perez J, Gonnelli A, Drouyer M, Chartier-Harlin MC, Taymans JM, Bubacco L, Greggio E, Hilfiker S. GTP binding regulates cellular localization of Parkinson's disease-associated LRRK2. Hum Mol Genet. 2017;26:2747–67.CrossRefPubMed Blanca Ramirez M, Lara Ordonez AJ, Fdez E, Madero-Perez J, Gonnelli A, Drouyer M, Chartier-Harlin MC, Taymans JM, Bubacco L, Greggio E, Hilfiker S. GTP binding regulates cellular localization of Parkinson's disease-associated LRRK2. Hum Mol Genet. 2017;26:2747–67.CrossRefPubMed
54.
go back to reference Dephoure N, Gould KL, Gygi SP, Kellogg DR. Mapping and analysis of phosphorylation sites: a quick guide for cell biologists. Mol Biol Cell. 2013;24:535–42.CrossRefPubMedPubMedCentral Dephoure N, Gould KL, Gygi SP, Kellogg DR. Mapping and analysis of phosphorylation sites: a quick guide for cell biologists. Mol Biol Cell. 2013;24:535–42.CrossRefPubMedPubMedCentral
55.
go back to reference Hunter T. Why nature chose phosphate to modify proteins. Philos Trans R Soc Lond Ser B Biol Sci. 2012;367:2513–6.CrossRef Hunter T. Why nature chose phosphate to modify proteins. Philos Trans R Soc Lond Ser B Biol Sci. 2012;367:2513–6.CrossRef
56.
go back to reference Pouthas F, Girard P, Lecaudey V, Ly TB, Gilmour D, Boulin C, Pepperkok R, Reynaud EG. In migrating cells, the Golgi complex and the position of the centrosome depend on geometrical constraints of the substratum. J Cell Sci. 2008;121:2406–14.CrossRefPubMed Pouthas F, Girard P, Lecaudey V, Ly TB, Gilmour D, Boulin C, Pepperkok R, Reynaud EG. In migrating cells, the Golgi complex and the position of the centrosome depend on geometrical constraints of the substratum. J Cell Sci. 2008;121:2406–14.CrossRefPubMed
57.
go back to reference Hattula K, Furuhjelm J, Arffman A, Peranen J. A Rab8-specific GDP/GTP exchange factor is involved in actin remodeling and polarized membrane transport. Mol Biol Cell. 2002;13:3268–80.CrossRefPubMedPubMedCentral Hattula K, Furuhjelm J, Arffman A, Peranen J. A Rab8-specific GDP/GTP exchange factor is involved in actin remodeling and polarized membrane transport. Mol Biol Cell. 2002;13:3268–80.CrossRefPubMedPubMedCentral
58.
go back to reference Villarroel-Campos D, Gastaldi L, Conde C, Caceres A, Gonzalez-Billault C. Rab-mediated trafficking role in neurite formation. J Neurochem. 2014;129:240–8.CrossRefPubMed Villarroel-Campos D, Gastaldi L, Conde C, Caceres A, Gonzalez-Billault C. Rab-mediated trafficking role in neurite formation. J Neurochem. 2014;129:240–8.CrossRefPubMed
59.
go back to reference Lai YC, Kondapalli C, Lehneck R, Procter JB, Dill BD, Woodroof HI, Gourlay R, Peggie M, Macartney TJ, Corti O, et al. Phosphoproteomic screening identifies Rab GTPases as novel downstream targets of PINK1. EMBO J. 2015;34:2840–61.CrossRefPubMedPubMedCentral Lai YC, Kondapalli C, Lehneck R, Procter JB, Dill BD, Woodroof HI, Gourlay R, Peggie M, Macartney TJ, Corti O, et al. Phosphoproteomic screening identifies Rab GTPases as novel downstream targets of PINK1. EMBO J. 2015;34:2840–61.CrossRefPubMedPubMedCentral
60.
go back to reference Gitler AD, Bevis BJ, Shorter J, Strathearn KE, Hamamichi S, Su LJ, Caldwell KA, Caldwell GA, Rochet JC, McCaffery JM, et al. The Parkinson's disease protein alpha-synuclein disrupts cellular Rab homeostasis. Proc Natl Acad Sci U S A. 2008;105:145–50.CrossRefPubMed Gitler AD, Bevis BJ, Shorter J, Strathearn KE, Hamamichi S, Su LJ, Caldwell KA, Caldwell GA, Rochet JC, McCaffery JM, et al. The Parkinson's disease protein alpha-synuclein disrupts cellular Rab homeostasis. Proc Natl Acad Sci U S A. 2008;105:145–50.CrossRefPubMed
61.
go back to reference Agalliu I, San Luciano M, Mirelman A, Giladi N, Waro B, Aasly J, Inzelberg R, Hassin-Baer S, Friedman E, Ruiz-Martinez J, et al. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurol. 2015;72:58–65.CrossRefPubMedPubMedCentral Agalliu I, San Luciano M, Mirelman A, Giladi N, Waro B, Aasly J, Inzelberg R, Hassin-Baer S, Friedman E, Ruiz-Martinez J, et al. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurol. 2015;72:58–65.CrossRefPubMedPubMedCentral
62.
go back to reference Pellegrini L, Wetzel A, Granno S, Heaton G, Harvey K. Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci. 2017;74:409–34.CrossRefPubMed Pellegrini L, Wetzel A, Granno S, Heaton G, Harvey K. Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci. 2017;74:409–34.CrossRefPubMed
63.
go back to reference Soukup SF, Kuenen S, Vanhauwaert R, Manetsberger J, Hernandez-Diaz S, Swerts J, Schoovaerts N, Vilain S, Gounko NV, Vints K, et al. A LRRK2-dependent EndophilinA Phosphoswitch is critical for macroautophagy at presynaptic terminals. Neuron. 2016;92:829–44.CrossRefPubMed Soukup SF, Kuenen S, Vanhauwaert R, Manetsberger J, Hernandez-Diaz S, Swerts J, Schoovaerts N, Vilain S, Gounko NV, Vints K, et al. A LRRK2-dependent EndophilinA Phosphoswitch is critical for macroautophagy at presynaptic terminals. Neuron. 2016;92:829–44.CrossRefPubMed
64.
go back to reference Abeliovich A, Gitler AD. Defects in trafficking bridge Parkinson's disease pathology and genetics. Nature. 2016;539:207–16.CrossRefPubMed Abeliovich A, Gitler AD. Defects in trafficking bridge Parkinson's disease pathology and genetics. Nature. 2016;539:207–16.CrossRefPubMed
Metadata
Title
Parkinson disease-associated mutations in LRRK2 cause centrosomal defects via Rab8a phosphorylation
Authors
Jesús Madero-Pérez
Elena Fdez
Belén Fernández
Antonio J. Lara Ordóñez
Marian Blanca Ramírez
Patricia Gómez-Suaga
Dieter Waschbüsch
Evy Lobbestael
Veerle Baekelandt
Angus C. Nairn
Javier Ruiz-Martínez
Ana Aiastui
Adolfo López de Munain
Pawel Lis
Thomas Comptdaer
Jean-Marc Taymans
Marie-Christine Chartier-Harlin
Alexandria Beilina
Adriano Gonnelli
Mark R. Cookson
Elisa Greggio
Sabine Hilfiker
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2018
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-018-0235-y

Other articles of this Issue 1/2018

Molecular Neurodegeneration 1/2018 Go to the issue