Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2017

Open Access 01-12-2017 | Research article

Pathogenic LRRK2 variants are gain-of-function mutations that enhance LRRK2-mediated repression of β-catenin signaling

Authors: Daniel C. Berwick, Behzad Javaheri, Andrea Wetzel, Mark Hopkinson, Jonathon Nixon-Abell, Simone Grannò, Andrew A. Pitsillides, Kirsten Harvey

Published in: Molecular Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Background

LRRK2 mutations and risk variants increase susceptibility to inherited and idiopathic Parkinson’s disease, while recent studies have identified potential protective variants. This, and the fact that LRRK2 mutation carriers develop symptoms and brain pathology almost indistinguishable from idiopathic Parkinson’s disease, has led to enormous interest in this protein. LRRK2 has been implicated in a range of cellular events, but key among them is canonical Wnt signalling, which results in increased levels of transcriptionally active β-catenin. This pathway is critical for the development and survival of the midbrain dopaminergic neurones typically lost in Parkinson’s disease.

Methods

Here we use Lrrk2 knockout mice and fibroblasts to investigate the effect of loss of Lrrk2 on canonical Wnt signalling in vitro and in vivo. Micro-computed tomography was used to study predicted tibial strength, while pulldown assays were employed to measure brain β-catenin levels. A combination of luciferase assays, immunofluorescence and co-immunoprecipitation were performed to measure canonical Wnt activity and investigate the relationship between LRRK2 and β-catenin. TOPflash assays are also used to study the effects of LRRK2 kinase inhibition and pathogenic and protective LRRK2 mutations on Wnt signalling. Data were tested by Analysis of Variance.

Results

Loss of Lrrk2 causes a dose-dependent increase in the levels of transcriptionally active β-catenin in the brain, and alters tibial bone architecture, decreasing the predicted risk of fracture. Lrrk2 knockout cells display increased TOPflash and Axin2 promoter activities, both basally and following Wnt activation. Consistently, over-expressed LRRK2 was found to bind β-catenin and repress TOPflash activation. Some pathogenic LRRK2 mutations and risk variants further suppressed TOPflash, whereas the protective R1398H variant increased Wnt signalling activity. LRRK2 kinase inhibitors affected canonical Wnt signalling differently due to off-targeting; however, specific LRRK2 inhibition reduced canonical Wnt signalling similarly to pathogenic mutations.

Conclusions

Loss of LRRK2 causes increased canonical Wnt activity in vitro and in vivo. In agreement, over-expressed LRRK2 binds and represses β-catenin, suggesting LRRK2 may act as part of the β-catenin destruction complex. Since some pathogenic LRRK2 mutations enhance this effect while the protective R1398H variant relieves it, our data strengthen the notion that decreased canonical Wnt activity is central to Parkinson’s disease pathogenesis.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Kumari U, Tan EK. LRRK2 in Parkinson’s disease: genetic and clinical studies from patients. FEBS J. 2009;276:6455–63.CrossRefPubMed Kumari U, Tan EK. LRRK2 in Parkinson’s disease: genetic and clinical studies from patients. FEBS J. 2009;276:6455–63.CrossRefPubMed
3.
go back to reference Parkinson J. An essay on the shaking palsy. 1817. J Neuropsychiatry Clin Neurosci. 2002;14:223–36.CrossRefPubMed Parkinson J. An essay on the shaking palsy. 1817. J Neuropsychiatry Clin Neurosci. 2002;14:223–36.CrossRefPubMed
4.
go back to reference Berwick DC, Harvey K. LRRK2 signaling pathways: the key to unlocking neurodegeneration? Trends Cell Biol. 2011;21:257–65.CrossRefPubMed Berwick DC, Harvey K. LRRK2 signaling pathways: the key to unlocking neurodegeneration? Trends Cell Biol. 2011;21:257–65.CrossRefPubMed
5.
go back to reference Greggio E, Cookson MR. Leucine-rich repeat kinase 2 mutations and Parkinson’s disease: three questions. ASN Neuro. 2009;1:1.CrossRef Greggio E, Cookson MR. Leucine-rich repeat kinase 2 mutations and Parkinson’s disease: three questions. ASN Neuro. 2009;1:1.CrossRef
6.
go back to reference Lewis PA, Manzoni C. LRRK2 and human disease: a complicated question or a question of complexes? Sci Signal. 2012;5:pe2.CrossRefPubMed Lewis PA, Manzoni C. LRRK2 and human disease: a complicated question or a question of complexes? Sci Signal. 2012;5:pe2.CrossRefPubMed
7.
8.
go back to reference Ho CC, Rideout HJ, Ribe E, Troy CM, Dauer WT. The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci. 2009;29:1011–6.CrossRefPubMedPubMedCentral Ho CC, Rideout HJ, Ribe E, Troy CM, Dauer WT. The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci. 2009;29:1011–6.CrossRefPubMedPubMedCentral
9.
go back to reference 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;12:1063–70.CrossRefPubMedPubMedCentral 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;12:1063–70.CrossRefPubMedPubMedCentral
10.
go back to reference Gardet A, Benita Y, Li C, Sands BE, Ballester I, Stevens C, Korzenik JR, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185:5577–85.CrossRefPubMedPubMedCentral Gardet A, Benita Y, Li C, Sands BE, Ballester I, Stevens C, Korzenik JR, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185:5577–85.CrossRefPubMedPubMedCentral
11.
go back to reference 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:e39132.CrossRefPubMedPubMedCentral 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:e39132.CrossRefPubMedPubMedCentral
12.
go back to reference Chia R, Haddock S, Beilina A, Rudenko IN, Mamais A, Kaganovich A, et al. Phosphorylation of LRRK2 by casein kinase 1α regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat Commun. 2014;5:5827.CrossRefPubMedPubMedCentral Chia R, Haddock S, Beilina A, Rudenko IN, Mamais A, Kaganovich A, et al. Phosphorylation of LRRK2 by casein kinase 1α regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat Commun. 2014;5:5827.CrossRefPubMedPubMedCentral
13.
go back to reference Häbig K, Walter M, Poths S, Riess O, Bonin M. RNA interference of LRRK2-microarray expression analysis of a Parkinson’s disease key player. Neurogenetics. 2008;9:83–94.CrossRefPubMed Häbig K, Walter M, Poths S, Riess O, Bonin M. RNA interference of LRRK2-microarray expression analysis of a Parkinson’s disease key player. Neurogenetics. 2008;9:83–94.CrossRefPubMed
14.
go back to reference Sancho RM, Law BM, Harvey K. Mutations in the LRRK2 Roc-COR tandem domain link Parkinson’s disease to Wnt signalling pathways. Hum Mol Genet. 2009;18:3955–68.CrossRefPubMedPubMedCentral Sancho RM, Law BM, Harvey K. Mutations in the LRRK2 Roc-COR tandem domain link Parkinson’s disease to Wnt signalling pathways. Hum Mol Genet. 2009;18:3955–68.CrossRefPubMedPubMedCentral
15.
go back to reference Lin CH, Tsai PI, Wu RM, Chien CT. LRRK2 G2019S mutation induces dendrite degeneration through mislocalization and phosphorylation of tau by recruiting autoactivated GSK3ß. J Neurosci. 2010;30:13138–49.CrossRefPubMed Lin CH, Tsai PI, Wu RM, Chien CT. LRRK2 G2019S mutation induces dendrite degeneration through mislocalization and phosphorylation of tau by recruiting autoactivated GSK3ß. J Neurosci. 2010;30:13138–49.CrossRefPubMed
16.
go back to reference Berwick DC, Harvey K. LRRK2 functions as a Wnt signaling scaffold, bridging cytosolic proteins and membrane-localized LRP6. Hum Mol Genet. 2012;21:4966–79.CrossRefPubMedPubMedCentral Berwick DC, Harvey K. LRRK2 functions as a Wnt signaling scaffold, bridging cytosolic proteins and membrane-localized LRP6. Hum Mol Genet. 2012;21:4966–79.CrossRefPubMedPubMedCentral
17.
go back to reference Dusonchet J, Li H, Guillily M, Liu M, Stafa K, Derada Troletti C, et al. Parkinson’s disease gene regulatory network identifies the signaling protein RGS2 as a modulator of LRRK2 activity and neuronal toxicity. Hum Mol Genet. 2014;23:4887–905.CrossRefPubMedPubMedCentral Dusonchet J, Li H, Guillily M, Liu M, Stafa K, Derada Troletti C, et al. Parkinson’s disease gene regulatory network identifies the signaling protein RGS2 as a modulator of LRRK2 activity and neuronal toxicity. Hum Mol Genet. 2014;23:4887–905.CrossRefPubMedPubMedCentral
18.
go back to reference Nixon-Abell J, Berwick DC, Grannó S, Spain VA, Blackstone C, Harvey K. Protective LRRK2 R1398H variant enhances GTPase and Wnt signalling activity. Front Mol Neurosci. 2016;9:18.CrossRefPubMedPubMedCentral Nixon-Abell J, Berwick DC, Grannó S, Spain VA, Blackstone C, Harvey K. Protective LRRK2 R1398H variant enhances GTPase and Wnt signalling activity. Front Mol Neurosci. 2016;9:18.CrossRefPubMedPubMedCentral
19.
go back to reference 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;5:e12813.CrossRefPubMedPubMedCentral 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;5:e12813.CrossRefPubMedPubMedCentral
22.
go back to reference van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205–14.CrossRefPubMed van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205–14.CrossRefPubMed
23.
go back to reference Atkinson JM, Rank KB, Zeng Y, Capen A, Yadav V, Manro JR, et al. Activating the Wnt/β-Catenin Pathway for the Treatment of Melanoma—Application of LY2090314, a Novel Selective Inhibitor of Glycogen Synthase Kinase-3. PLoS One. 2015;10:e0125028.CrossRefPubMedPubMedCentral Atkinson JM, Rank KB, Zeng Y, Capen A, Yadav V, Manro JR, et al. Activating the Wnt/β-Catenin Pathway for the Treatment of Melanoma—Application of LY2090314, a Novel Selective Inhibitor of Glycogen Synthase Kinase-3. PLoS One. 2015;10:e0125028.CrossRefPubMedPubMedCentral
24.
go back to reference Kawakami T, Ren S, Duffield JS. Wnt signalling in kidney diseases: dual roles in renal injury and repair. J Pathol. 2013;229:221–31.CrossRefPubMed Kawakami T, Ren S, Duffield JS. Wnt signalling in kidney diseases: dual roles in renal injury and repair. J Pathol. 2013;229:221–31.CrossRefPubMed
25.
go back to reference Cheng JH, She H, Han YP, Wang J, Xiong S, Asahina K, et al. Wnt antagonism inhibits hepatic stellate cell activation and liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2008;294:G39–49.CrossRefPubMed Cheng JH, She H, Han YP, Wang J, Xiong S, Asahina K, et al. Wnt antagonism inhibits hepatic stellate cell activation and liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2008;294:G39–49.CrossRefPubMed
26.
go back to reference Königshoff M, Balsara N, Pfaff EM, Kramer M, Chrobak I, Seeger W, et al. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS One. 2008;3:e2142.CrossRefPubMedPubMedCentral Königshoff M, Balsara N, Pfaff EM, Kramer M, Chrobak I, Seeger W, et al. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS One. 2008;3:e2142.CrossRefPubMedPubMedCentral
27.
go back to reference Berwick DC, Harvey K. The importance of Wnt signalling for neurodegeneration in Parkinson’s disease. Biochem Soc Trans. 2012;40:1123–8.CrossRefPubMed Berwick DC, Harvey K. The importance of Wnt signalling for neurodegeneration in Parkinson’s disease. Biochem Soc Trans. 2012;40:1123–8.CrossRefPubMed
28.
go back to reference Inestrosa NC, Montecinos-Oliva C, Fuenzalida M. Wnt signaling: role in Alzheimer disease and schizophrenia. J Neuroimmune Pharmacol. 2012;7:788–807.CrossRefPubMed Inestrosa NC, Montecinos-Oliva C, Fuenzalida M. Wnt signaling: role in Alzheimer disease and schizophrenia. J Neuroimmune Pharmacol. 2012;7:788–807.CrossRefPubMed
29.
go back to reference Voleti B, Duman RS. The roles of neurotrophic factor and Wnt signaling in depression. Clin Pharmacol Ther. 2012;91:333–8.CrossRefPubMed Voleti B, Duman RS. The roles of neurotrophic factor and Wnt signaling in depression. Clin Pharmacol Ther. 2012;91:333–8.CrossRefPubMed
30.
go back to reference Babij P, Zhao W, Small C, Kharode Y, Yaworsky PJ, Bouxsein ML, et al. High bone mass in mice expressing a mutant LRP5 gene. J Bone Miner Res. 2003;18:960–74.CrossRefPubMed Babij P, Zhao W, Small C, Kharode Y, Yaworsky PJ, Bouxsein ML, et al. High bone mass in mice expressing a mutant LRP5 gene. J Bone Miner Res. 2003;18:960–74.CrossRefPubMed
31.
go back to reference Bodine PV, Zhao W, Kharode YP, Bex FJ, Lambert AJ, Goad MB, et al. The Wnt antagonist secreted frizzled-related protein-1 is a negative regulator of trabecular bone formation in adult mice. Mol Endocrinol. 2004;18:1222–37.CrossRefPubMed Bodine PV, Zhao W, Kharode YP, Bex FJ, Lambert AJ, Goad MB, et al. The Wnt antagonist secreted frizzled-related protein-1 is a negative regulator of trabecular bone formation in adult mice. Mol Endocrinol. 2004;18:1222–37.CrossRefPubMed
32.
go back to reference Boyden LM, Mao J, Belsky J, Mitzner L, Farh A, Mitnick MA, et al. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med. 2002;346:1513–21.CrossRefPubMed Boyden LM, Mao J, Belsky J, Mitzner L, Farh A, Mitnick MA, et al. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med. 2002;346:1513–21.CrossRefPubMed
33.
go back to reference Johnson ML, Gong G, Kimberling W, Recker SM, Kimmel DB, Recker RR. Linkage of a Gene Causing High Bone Mass to Human Chromosome 11 (11q12-13). Am J Hum Genet. 1997;60:1326–32.CrossRefPubMedPubMedCentral Johnson ML, Gong G, Kimberling W, Recker SM, Kimmel DB, Recker RR. Linkage of a Gene Causing High Bone Mass to Human Chromosome 11 (11q12-13). Am J Hum Genet. 1997;60:1326–32.CrossRefPubMedPubMedCentral
34.
go back to reference Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, et al. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet. 2002;70:11–9.CrossRefPubMed Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, et al. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet. 2002;70:11–9.CrossRefPubMed
35.
go back to reference Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, et al. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am J Hum Genet. 2003;72:763–71.CrossRefPubMedPubMedCentral Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, et al. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am J Hum Genet. 2003;72:763–71.CrossRefPubMedPubMedCentral
36.
go back to reference Glass 2nd DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–64.CrossRefPubMed Glass 2nd DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–64.CrossRefPubMed
37.
go back to reference Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–23.CrossRefPubMed Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–23.CrossRefPubMed
38.
go back to reference Gong Y, Vikkula M, Boon L, Liu J, Beighton P, Ramesar R, et al. Osteoporosis-pseudoglioma syndrome, a disorder affecting skeletal strength and vision, is assigned to chromosome region 11q12-13. Am J Hum Genet. 1996;59:146–51.PubMedPubMedCentral Gong Y, Vikkula M, Boon L, Liu J, Beighton P, Ramesar R, et al. Osteoporosis-pseudoglioma syndrome, a disorder affecting skeletal strength and vision, is assigned to chromosome region 11q12-13. Am J Hum Genet. 1996;59:146–51.PubMedPubMedCentral
39.
go back to reference Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, et al. Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005;280:21162–8.CrossRefPubMed Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, et al. Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005;280:21162–8.CrossRefPubMed
40.
go back to reference Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass 2nd DA, et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–14.CrossRefPubMedPubMedCentral Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass 2nd DA, et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–14.CrossRefPubMedPubMedCentral
41.
go back to reference Javaheri B, Stern AR, Lara N, Dallas M, Zhao H, Liu Y, et al. Deletion of a single beta-catenin allele in osteocytes abolishes the bone anabolic response to loading. J Bone Miner Res. 2014;29:705–15.CrossRefPubMedPubMedCentral Javaheri B, Stern AR, Lara N, Dallas M, Zhao H, Liu Y, et al. Deletion of a single beta-catenin allele in osteocytes abolishes the bone anabolic response to loading. J Bone Miner Res. 2014;29:705–15.CrossRefPubMedPubMedCentral
42.
go back to reference Parisiadou L, Xie C, Cho HJ, Lin X, Gu XL, Long CX, et al. 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, et al. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci. 2009;29:13971–80.CrossRefPubMedPubMedCentral
43.
go back to reference Javaheri B, Carriero A, Staines KA, Chang YM, Houston DA, Oldknow KJ, et al. Phospho1 deficiency transiently modifies bone architecture yet produces consistent modification in osteocyte differentiation and vascular porosity with ageing. Bone. 2015;81:277–91.CrossRefPubMedPubMedCentral Javaheri B, Carriero A, Staines KA, Chang YM, Houston DA, Oldknow KJ, et al. Phospho1 deficiency transiently modifies bone architecture yet produces consistent modification in osteocyte differentiation and vascular porosity with ageing. Bone. 2015;81:277–91.CrossRefPubMedPubMedCentral
44.
go back to reference Doube M, Klosowski MM, Arganda-Carreras I, Cordelieres FP, Dougherty RP, Jackson JS, et al. BoneJ: Free and extensible bone image analysis in ImageJ. Bone. 2010;47:1076–9.CrossRefPubMedPubMedCentral Doube M, Klosowski MM, Arganda-Carreras I, Cordelieres FP, Dougherty RP, Jackson JS, et al. BoneJ: Free and extensible bone image analysis in ImageJ. Bone. 2010;47:1076–9.CrossRefPubMedPubMedCentral
45.
go back to reference Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5.CrossRefPubMed Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5.CrossRefPubMed
46.
go back to reference Luckert K, Götschel F, Sorger PK, Hecht A, Joos TO, Pötz O. Snapshots of protein dynamics and post-translational modifications in one experiment—beta catenin and its functions. Mol Cell Proteomics. 2011;10:M110.007377.CrossRefPubMedPubMedCentral Luckert K, Götschel F, Sorger PK, Hecht A, Joos TO, Pötz O. Snapshots of protein dynamics and post-translational modifications in one experiment—beta catenin and its functions. Mol Cell Proteomics. 2011;10:M110.007377.CrossRefPubMedPubMedCentral
47.
go back to reference Kolligs FT, Hu G, Dang CV, Fearon ER. Neoplastic transformation of RK3E by mutant beta-catenin requires deregulation of Tcf/Lef transcription but not activation of c-myc expression. Mol Cell Biol. 1999;19:5696–706.CrossRefPubMedPubMedCentral Kolligs FT, Hu G, Dang CV, Fearon ER. Neoplastic transformation of RK3E by mutant beta-catenin requires deregulation of Tcf/Lef transcription but not activation of c-myc expression. Mol Cell Biol. 1999;19:5696–706.CrossRefPubMedPubMedCentral
48.
go back to reference Jho EH, Zhang T, Domon C, Joo CK, Freund JN, Costantini F. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 2002;22:1172–83.CrossRefPubMedPubMedCentral Jho EH, Zhang T, Domon C, Joo CK, Freund JN, Costantini F. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 2002;22:1172–83.CrossRefPubMedPubMedCentral
49.
go back to reference Rivers A, Gietzen KF, Vielhaber E, Virshup DM. Regulation of casein kinase Iepsilon and casein kinase I delta by an in vivo futile phosphorylation cycle. J Biol Chem. 1998;273:15980–4.CrossRefPubMed Rivers A, Gietzen KF, Vielhaber E, Virshup DM. Regulation of casein kinase Iepsilon and casein kinase I delta by an in vivo futile phosphorylation cycle. J Biol Chem. 1998;273:15980–4.CrossRefPubMed
50.
go back to reference He X, Saint-Jeannet JP, Woodgett JR, Varmus HE, Dawid IB. Glycogne synthase kinase-3 and dorsoventral patterning in Xenopus embryos. Nature. 1995;374:617–22.CrossRefPubMed He X, Saint-Jeannet JP, Woodgett JR, Varmus HE, Dawid IB. Glycogne synthase kinase-3 and dorsoventral patterning in Xenopus embryos. Nature. 1995;374:617–22.CrossRefPubMed
51.
go back to reference Law BM, Spain VA, Leinster VH, Chia R, Beilina A, Cho HJ, et al. A direct interaction between leucine-rich repeat kinase 2 and specific β-tubulin isoforms regulates tubulin acetylation. J Biol Chem. 2014;289:895–908.CrossRefPubMed Law BM, Spain VA, Leinster VH, Chia R, Beilina A, Cho HJ, et al. A direct interaction between leucine-rich repeat kinase 2 and specific β-tubulin isoforms regulates tubulin acetylation. J Biol Chem. 2014;289:895–908.CrossRefPubMed
52.
go back to reference 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;7:203–5.CrossRefPubMedPubMedCentral 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;7:203–5.CrossRefPubMedPubMedCentral
53.
54.
go back to reference Ramsden N, Perrin J, Ren Z, Lee BD, Zinn N, Dawson VL, et al. Chemoproteomics-based sign of potent LRRK2-selective lead compounds that attenuate Parkinson’s disease-related toxicity in human neurons. ACS Chem Biol. 2011;6:1021–8.CrossRefPubMedPubMedCentral Ramsden N, Perrin J, Ren Z, Lee BD, Zinn N, Dawson VL, et al. Chemoproteomics-based sign of potent LRRK2-selective lead compounds that attenuate Parkinson’s disease-related toxicity in human neurons. ACS Chem Biol. 2011;6:1021–8.CrossRefPubMedPubMedCentral
55.
go back to reference Xing W, Liu J, Cheng S, Vogel P, Mohan S, Brommage R. Targeted disruption of leucine-rich repeat kinase 1 but not leucine-rich repeat kinase 2 in mice causes severe osteopetrosis. J Bone Miner Res. 2013;28:1962–74.CrossRefPubMed Xing W, Liu J, Cheng S, Vogel P, Mohan S, Brommage R. Targeted disruption of leucine-rich repeat kinase 1 but not leucine-rich repeat kinase 2 in mice causes severe osteopetrosis. J Bone Miner Res. 2013;28:1962–74.CrossRefPubMed
56.
go back to reference Veeman MT, Slusarski DC, Kaykas A, Louie SH, Moon RT. Zebrafish prickle, a modulator of noncanonical Wnt/Fz signaling, regulates gastrulation movements. Curr Biol. 2003;13:680–5.CrossRefPubMed Veeman MT, Slusarski DC, Kaykas A, Louie SH, Moon RT. Zebrafish prickle, a modulator of noncanonical Wnt/Fz signaling, regulates gastrulation movements. Curr Biol. 2003;13:680–5.CrossRefPubMed
57.
58.
go back to reference Faux MC, Coates JL, Catimel B, Cody S, Clayton AH, Layton MJ, et al. Recruitment of adenomatous polyposis coli and β-catenin to axin-puncta. Oncogene. 2008;27:5808–20.CrossRefPubMed Faux MC, Coates JL, Catimel B, Cody S, Clayton AH, Layton MJ, et al. Recruitment of adenomatous polyposis coli and β-catenin to axin-puncta. Oncogene. 2008;27:5808–20.CrossRefPubMed
59.
go back to reference Pronobis MI, Rusan NM, Peifer M. A novel GSK3-regulated APC:Axin interaction regulates Wnt signaling by driving a catalytic cycle of efficient βcatenin destruction. Elife. 2015;4:e08022.CrossRefPubMedPubMedCentral Pronobis MI, Rusan NM, Peifer M. A novel GSK3-regulated APC:Axin interaction regulates Wnt signaling by driving a catalytic cycle of efficient βcatenin destruction. Elife. 2015;4:e08022.CrossRefPubMedPubMedCentral
60.
go back to reference Swarup S, Pradhan-Sundd T, Verheyen EM. Genome-wide identification of phospho-regulators of Wnt signaling in Drospohila. Development. 2015;142:1502–15.CrossRefPubMed Swarup S, Pradhan-Sundd T, Verheyen EM. Genome-wide identification of phospho-regulators of Wnt signaling in Drospohila. Development. 2015;142:1502–15.CrossRefPubMed
61.
go back to reference Prasad BC, Clark SG. Wnt signaling establishes anteroposterior neuronal polarity and requires retromer in C. elegans. Development. 2006;133:1757–66.CrossRefPubMed Prasad BC, Clark SG. Wnt signaling establishes anteroposterior neuronal polarity and requires retromer in C. elegans. Development. 2006;133:1757–66.CrossRefPubMed
62.
go back to reference Anichtchik O, Diekmann H, Fleming A, Roach A, Goldsmith P, Rubinsztein DC. Loss of PINK1 function affects development and results in neurodegeneration in zebrafish. J Neurosci. 2008;28:8199–207.CrossRefPubMed Anichtchik O, Diekmann H, Fleming A, Roach A, Goldsmith P, Rubinsztein DC. Loss of PINK1 function affects development and results in neurodegeneration in zebrafish. J Neurosci. 2008;28:8199–207.CrossRefPubMed
63.
go back to reference Bheda A, Yue W, Gullapalli A, Whitehurst C, Liu R, Pagano JS, et al. Positive reciprocal regulation of ubiquitin C-terminal hydrolase L1 and beta-catenin/TCF signaling. PLoS One. 2009;4:e5955.CrossRefPubMedPubMedCentral Bheda A, Yue W, Gullapalli A, Whitehurst C, Liu R, Pagano JS, et al. Positive reciprocal regulation of ubiquitin C-terminal hydrolase L1 and beta-catenin/TCF signaling. PLoS One. 2009;4:e5955.CrossRefPubMedPubMedCentral
64.
go back to reference Rawal N, Corti O, Sacchetti P, Ardilla-Osorio H, Sehat B, Brice A, et al. Parkin protects dopaminergic neurons from excessive Wnt/beta-catenin signaling. Biochem Biophys Res Commun. 2009;388:473–8.CrossRefPubMed Rawal N, Corti O, Sacchetti P, Ardilla-Osorio H, Sehat B, Brice A, et al. Parkin protects dopaminergic neurons from excessive Wnt/beta-catenin signaling. Biochem Biophys Res Commun. 2009;388:473–8.CrossRefPubMed
65.
go back to reference Cruciat CM, Ohkawara B, Acebron SP, Karaulanov E, Reinhard C, Ingelfinger D, et al. Requirement of prorenin receptor and vacuolar H + −ATPase-mediated acidification for Wnt signaling. Science. 2010;327:459–63.CrossRefPubMed Cruciat CM, Ohkawara B, Acebron SP, Karaulanov E, Reinhard C, Ingelfinger D, et al. Requirement of prorenin receptor and vacuolar H + −ATPase-mediated acidification for Wnt signaling. Science. 2010;327:459–63.CrossRefPubMed
66.
go back to reference Zancan I, Bellesso S, Costa R, Salvalaio M, Stroppiano M, Hammond C, et al. Glucocerebrosidase deficiency in zebrafish affects primary bone ossification through increased oxidative stress and reduced Wnt/β-catenin signaling. Hum Mol Genet. 2015;24:1280–94.CrossRefPubMed Zancan I, Bellesso S, Costa R, Salvalaio M, Stroppiano M, Hammond C, et al. Glucocerebrosidase deficiency in zebrafish affects primary bone ossification through increased oxidative stress and reduced Wnt/β-catenin signaling. Hum Mol Genet. 2015;24:1280–94.CrossRefPubMed
67.
go back to reference Kwok JB, Hallupp M, Loy CT, Chan DK, Woo J, Mellick GD, et al. GSK3B polymorphisms alter transcription and splicing in Parkinson’s disease. Ann Neurol. 2005;58:829–39.CrossRefPubMed Kwok JB, Hallupp M, Loy CT, Chan DK, Woo J, Mellick GD, et al. GSK3B polymorphisms alter transcription and splicing in Parkinson’s disease. Ann Neurol. 2005;58:829–39.CrossRefPubMed
68.
go back to reference Arenas E. Wnt signaling in midbrain dopaminergic neuron development and regenerative medicine for Parkinson’s disease. J Mol Cell Biol. 2014;6:42–53.CrossRefPubMed Arenas E. Wnt signaling in midbrain dopaminergic neuron development and regenerative medicine for Parkinson’s disease. J Mol Cell Biol. 2014;6:42–53.CrossRefPubMed
69.
go back to reference Joksimovic M, Awatramani R. Wnt/β-catenin signaling in midbrain dopaminergic neuron specification and neurogenesis. J Mol Cell Biol. 2014;6:27–33.CrossRefPubMed Joksimovic M, Awatramani R. Wnt/β-catenin signaling in midbrain dopaminergic neuron specification and neurogenesis. J Mol Cell Biol. 2014;6:27–33.CrossRefPubMed
71.
go back to reference Inestrosa NC, Arenas E. Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci. 2010;11:77–86.CrossRefPubMed Inestrosa NC, Arenas E. Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci. 2010;11:77–86.CrossRefPubMed
73.
74.
go back to reference Cantuti-Castelvetri I, Keller-McGandy C, Bouzou B, Asteris G, Clark TW, Frosch MP, et al. Effects of gender on nigral gene expression and parkinson disease. Neurobiol Dis. 2007;26:606–14.CrossRefPubMedPubMedCentral Cantuti-Castelvetri I, Keller-McGandy C, Bouzou B, Asteris G, Clark TW, Frosch MP, et al. Effects of gender on nigral gene expression and parkinson disease. Neurobiol Dis. 2007;26:606–14.CrossRefPubMedPubMedCentral
75.
go back to reference L’Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Cossetti C, et al. Reactive astrocytes and Wnt/β-catenin signaling link nigrostriatal injury to repair in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Neurobiol Dis. 2011;41:508–27.CrossRefPubMed L’Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Cossetti C, et al. Reactive astrocytes and Wnt/β-catenin signaling link nigrostriatal injury to repair in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Neurobiol Dis. 2011;41:508–27.CrossRefPubMed
76.
go back to reference Gollamudi S, Johri A, Calingasan NY, Yang L, Elemento O, Beal MF. Concordant signaling pathways produced by pesticide exposure in mice correspond to pathways identified in human Parkinson’s disease. PLoS ONE. 2012;7:e36191.CrossRefPubMedPubMedCentral Gollamudi S, Johri A, Calingasan NY, Yang L, Elemento O, Beal MF. Concordant signaling pathways produced by pesticide exposure in mice correspond to pathways identified in human Parkinson’s disease. PLoS ONE. 2012;7:e36191.CrossRefPubMedPubMedCentral
78.
go back to reference Armstrong VJ, Muzylak M, Sunters A, Zaman G, Saxon LK, Price JS, et al. Wnt/beta-catenin signaling is a component of osteoblastic bone cell early responses to load-bearing and requires estrogen receptor alpha. J Biol Chem. 2007;282:20715–27.CrossRefPubMed Armstrong VJ, Muzylak M, Sunters A, Zaman G, Saxon LK, Price JS, et al. Wnt/beta-catenin signaling is a component of osteoblastic bone cell early responses to load-bearing and requires estrogen receptor alpha. J Biol Chem. 2007;282:20715–27.CrossRefPubMed
79.
go back to reference Bex F, Green P, Marzolf J, Babij P, Yaworsky P, Kharode Y. The human LRP5 G171V mutation in mice alters the skeletal response to limb unloading but not to ovariectomy. J Bone Miner Res. 2003;18:S60. Bex F, Green P, Marzolf J, Babij P, Yaworsky P, Kharode Y. The human LRP5 G171V mutation in mice alters the skeletal response to limb unloading but not to ovariectomy. J Bone Miner Res. 2003;18:S60.
80.
go back to reference Johnson ML, Harnish K, Nusse R, Van Hul W. LRP5 and Wnt signaling: a union made for bone. J Bone Miner Res. 2004;19:1749–57.CrossRefPubMed Johnson ML, Harnish K, Nusse R, Van Hul W. LRP5 and Wnt signaling: a union made for bone. J Bone Miner Res. 2004;19:1749–57.CrossRefPubMed
81.
go back to reference Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, Cullen DM, Zhao W, Li C, et al. Wnt/beta-catenin signaling is a normal physiological response to mechanical loading in bone. J Biol Chem. 2006;281:31720–8.CrossRefPubMed Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, Cullen DM, Zhao W, Li C, et al. Wnt/beta-catenin signaling is a normal physiological response to mechanical loading in bone. J Biol Chem. 2006;281:31720–8.CrossRefPubMed
82.
go back to reference Sawakami K, Robling AG, Ai M, Pitner ND, Liu D, Warden SJ, et al. The Wnt co-receptor LRP5 is essential for skeletal mechanotransduction but not for the anabolic bone response to parathyroid hormone treatment. J Biol Chem. 2006;281:23698–711.CrossRefPubMed Sawakami K, Robling AG, Ai M, Pitner ND, Liu D, Warden SJ, et al. The Wnt co-receptor LRP5 is essential for skeletal mechanotransduction but not for the anabolic bone response to parathyroid hormone treatment. J Biol Chem. 2006;281:23698–711.CrossRefPubMed
83.
go back to reference Galceran J, Farinas I, Depew MJ, Clevers H, Grosschedl R. Wnt3a−/−−like phenotype and limb deficiency in Lef1(−/−)Tcf1(−/−) mice. Genes Dev. 1999;13:709–17.CrossRefPubMedPubMedCentral Galceran J, Farinas I, Depew MJ, Clevers H, Grosschedl R. Wnt3a−/−−like phenotype and limb deficiency in Lef1(−/−)Tcf1(−/−) mice. Genes Dev. 1999;13:709–17.CrossRefPubMedPubMedCentral
84.
go back to reference Kelly OG, Pinson KI, Skarnes WC. The Wnt co-receptors Lrp5 and Lrp6 are essential for gastrulation in mice. Development. 2004;131:2803–15.CrossRefPubMed Kelly OG, Pinson KI, Skarnes WC. The Wnt co-receptors Lrp5 and Lrp6 are essential for gastrulation in mice. Development. 2004;131:2803–15.CrossRefPubMed
85.
go back to reference Ikeya M, Lee SM, Johnson JE, McMahon AP, Takada S. Wnt signalling required for expansion of neural crest and CNS progenitors. Nature. 1997;389:966–70.CrossRefPubMed Ikeya M, Lee SM, Johnson JE, McMahon AP, Takada S. Wnt signalling required for expansion of neural crest and CNS progenitors. Nature. 1997;389:966–70.CrossRefPubMed
86.
go back to reference Ikeya M, Takada S. Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome. Development. 1998;125:4969–76.PubMed Ikeya M, Takada S. Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome. Development. 1998;125:4969–76.PubMed
87.
go back to reference Lijam N, Paylor R, McDonald MP, Crawley JN, Deng CX, Herrup K, et al. Social interaction and sensorimotor gating abnormalities in mice lacking Dvl1. Cell. 1997;90:895–905.CrossRefPubMed Lijam N, Paylor R, McDonald MP, Crawley JN, Deng CX, Herrup K, et al. Social interaction and sensorimotor gating abnormalities in mice lacking Dvl1. Cell. 1997;90:895–905.CrossRefPubMed
88.
go back to reference Hamblet NS, Lijam N, Ruiz-Lozano P, Wang J, Yang Y, Luo Z, et al. Dishevelled 2 is essential for cardiac outflow tract development, somite segmentation and neural tube closure. Development. 2002;129:5827–38.CrossRefPubMed Hamblet NS, Lijam N, Ruiz-Lozano P, Wang J, Yang Y, Luo Z, et al. Dishevelled 2 is essential for cardiac outflow tract development, somite segmentation and neural tube closure. Development. 2002;129:5827–38.CrossRefPubMed
89.
go back to reference Ellwanger K, Saito H, Clement-Lacroix P, Maltry N, Niedermeyer J, Lee WK, et al. Targeted disruption of the Wnt regulator Kremen induces limb defects and high bone density. Mol Cell Biol. 2008;28:4875–82.CrossRefPubMedPubMedCentral Ellwanger K, Saito H, Clement-Lacroix P, Maltry N, Niedermeyer J, Lee WK, et al. Targeted disruption of the Wnt regulator Kremen induces limb defects and high bone density. Mol Cell Biol. 2008;28:4875–82.CrossRefPubMedPubMedCentral
91.
go back to reference Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher 3rd RJ, 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;107:9879–84.CrossRefPubMedPubMedCentral Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher 3rd RJ, 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;107:9879–84.CrossRefPubMedPubMedCentral
92.
go back to reference Ness D, Ren Z, Gardai S, Sharpnack D, Johnson VJ, Brennan RJ, 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.CrossRefPubMedPubMedCentral Ness D, Ren Z, Gardai S, Sharpnack D, Johnson VJ, Brennan RJ, 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.CrossRefPubMedPubMedCentral
93.
go back to reference 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:e80705.CrossRefPubMedPubMedCentral 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:e80705.CrossRefPubMedPubMedCentral
94.
go back to reference Guo Y, Xiao L, Sun L, Liu F. Wnt/beta-catenin signaling: a promising new target for fibrosis diseases. Physiol Res. 2012;61:337–46.PubMed Guo Y, Xiao L, Sun L, Liu F. Wnt/beta-catenin signaling: a promising new target for fibrosis diseases. Physiol Res. 2012;61:337–46.PubMed
95.
go back to reference Dennison EM, Compston JE, Flahive J, Siris ES, Gehlbach SH, Adachi JD, et al. Effect of co-morbidities on fracture risk: findings from the Global Longitudinal Study of Osteoporosis in Women (GLOW). Bone. 2012;50:1288–93.CrossRefPubMedPubMedCentral Dennison EM, Compston JE, Flahive J, Siris ES, Gehlbach SH, Adachi JD, et al. Effect of co-morbidities on fracture risk: findings from the Global Longitudinal Study of Osteoporosis in Women (GLOW). Bone. 2012;50:1288–93.CrossRefPubMedPubMedCentral
96.
go back to reference Invernizzi M, Carda S, Viscontini GS, Cisari C. Osteoporosis in Parkinson’s disease. Parkinsonism Relat Disord. 2009;15:339–46.CrossRefPubMed Invernizzi M, Carda S, Viscontini GS, Cisari C. Osteoporosis in Parkinson’s disease. Parkinsonism Relat Disord. 2009;15:339–46.CrossRefPubMed
97.
go back to reference Torsney KM, Noyce AJ, Doherty KM, Bestwick JP, Dobson R, Lees AJ. Bone health in Parkinson’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2014;85:1159–66.CrossRefPubMedPubMedCentral Torsney KM, Noyce AJ, Doherty KM, Bestwick JP, Dobson R, Lees AJ. Bone health in Parkinson’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2014;85:1159–66.CrossRefPubMedPubMedCentral
Metadata
Title
Pathogenic LRRK2 variants are gain-of-function mutations that enhance LRRK2-mediated repression of β-catenin signaling
Authors
Daniel C. Berwick
Behzad Javaheri
Andrea Wetzel
Mark Hopkinson
Jonathon Nixon-Abell
Simone Grannò
Andrew A. Pitsillides
Kirsten Harvey
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2017
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-017-0153-4

Other articles of this Issue 1/2017

Molecular Neurodegeneration 1/2017 Go to the issue