1
|
Rinaldi C, Waters CS, Li Z, Kumbier K, Rao L, Nichols RJ, Jacobson MP, Wu LF, Altschuler SJ. Dissecting the effects of GTPase and kinase domain mutations on LRRK2 endosomal localization and activity. Cell Rep 2023; 42:112447. [PMID: 37141099 DOI: 10.1016/j.celrep.2023.112447] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/21/2023] [Accepted: 04/13/2023] [Indexed: 05/05/2023] Open
Abstract
Parkinson's disease-causing leucine-rich repeat kinase 2 (LRRK2) mutations lead to varying degrees of Rab GTPase hyperphosphorylation. Puzzlingly, LRRK2 GTPase-inactivating mutations-which do not affect intrinsic kinase activity-lead to higher levels of cellular Rab phosphorylation than kinase-activating mutations. Here, we investigate whether mutation-dependent differences in LRRK2 cellular localization could explain this discrepancy. We discover that blocking endosomal maturation leads to the rapid formation of mutant LRRK2+ endosomes on which LRRK2 phosphorylates substrate Rabs. LRRK2+ endosomes are maintained through positive feedback, which mutually reinforces membrane localization of LRRK2 and phosphorylated Rab substrates. Furthermore, across a panel of mutants, cells expressing GTPase-inactivating mutants form strikingly more LRRK2+ endosomes than cells expressing kinase-activating mutants, resulting in higher total cellular levels of phosphorylated Rabs. Our study suggests that the increased probability that LRRK2 GTPase-inactivating mutants are retained on intracellular membranes compared to kinase-activating mutants leads to higher substrate phosphorylation.
Collapse
Affiliation(s)
- Capria Rinaldi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christopher S Waters
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Zizheng Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Karl Kumbier
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lee Rao
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - R Jeremy Nichols
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Steven J Altschuler
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
2
|
Weindel CG, Martinez EL, Zhao X, Mabry CJ, Bell SL, Vail KJ, Coleman AK, VanPortfliet JJ, Zhao B, Wagner AR, Azam S, Scott HM, Li P, West AP, Karpac J, Patrick KL, Watson RO. Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell 2022; 185:3214-3231.e23. [PMID: 35907404 PMCID: PMC9531054 DOI: 10.1016/j.cell.2022.06.038] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/05/2022] [Accepted: 06/18/2022] [Indexed: 10/16/2022]
Abstract
Although mutations in mitochondrial-associated genes are linked to inflammation and susceptibility to infection, their mechanistic contributions to immune outcomes remain ill-defined. We discovered that the disease-associated gain-of-function allele Lrrk2G2019S (leucine-rich repeat kinase 2) perturbs mitochondrial homeostasis and reprograms cell death pathways in macrophages. When the inflammasome is activated in Lrrk2G2019S macrophages, elevated mitochondrial ROS (mtROS) directs association of the pore-forming protein gasdermin D (GSDMD) to mitochondrial membranes. Mitochondrial GSDMD pore formation then releases mtROS, promoting a switch to RIPK1/RIPK3/MLKL-dependent necroptosis. Consistent with enhanced necroptosis, infection of Lrrk2G2019S mice with Mycobacterium tuberculosis elicits hyperinflammation and severe immunopathology. Our findings suggest a pivotal role for GSDMD as an executer of multiple cell death pathways and demonstrate that mitochondrial dysfunction can direct immune outcomes via cell death modality switching. This work provides insights into how LRRK2 mutations manifest or exacerbate human diseases and identifies GSDMD-dependent necroptosis as a potential target to limit Lrrk2G2019S-mediated immunopathology.
Collapse
Affiliation(s)
- Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Eduardo L Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Xiao Zhao
- Department of Molecular and Cellular Medicine, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Cory J Mabry
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA; Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Krystal J Vail
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Aja K Coleman
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Jordyn J VanPortfliet
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Baoyu Zhao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Allison R Wagner
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Sikandar Azam
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Haley M Scott
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA.
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
3
|
Marchand A, Sarchione A, Athanasopoulos PS, Roy HBL, Goveas L, Magnez R, Drouyer M, Emanuele M, Ho FY, Liberelle M, Melnyk P, Lebègue N, Thuru X, Nichols RJ, Greggio E, Kortholt A, Galli T, Chartier-Harlin MC, Taymans JM. A Phosphosite Mutant Approach on LRRK2 Links Phosphorylation and Dephosphorylation to Protective and Deleterious Markers, Respectively. Cells 2022; 11:cells11061018. [PMID: 35326469 PMCID: PMC8946913 DOI: 10.3390/cells11061018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/23/2022] Open
Abstract
The Leucine Rich Repeat Kinase 2 (LRRK2) gene is a major genetic determinant of Parkinson’s disease (PD), encoding a homonymous multi-domain protein with two catalytic activities, GTPase and Kinase, involved in intracellular signaling and trafficking. LRRK2 is phosphorylated at multiple sites, including a cluster of autophosphorylation sites in the GTPase domain and a cluster of heterologous phosphorylation sites at residues 860 to 976. Phosphorylation at these latter sites is found to be modified in brains of PD patients, as well as for some disease mutant forms of LRRK2. The main aim of this study is to investigate the functional consequences of LRRK2 phosphorylation or dephosphorylation at LRRK2’s heterologous phosphorylation sites. To this end, we generated LRRK2 phosphorylation site mutants and studied how these affected LRRK2 catalytic activity, neurite outgrowth and lysosomal physiology in cellular models. We show that phosphorylation of RAB8a and RAB10 substrates are reduced with phosphomimicking forms of LRRK2, while RAB29 induced activation of LRRK2 kinase activity is enhanced for phosphodead forms of LRRK2. Considering the hypothesis that PD pathology is associated to increased LRRK2 kinase activity, our results suggest that for its heterologous phosphorylation sites LRRK2 phosphorylation correlates to healthy phenotypes and LRRK2 dephosphorylation correlates to phenotypes associated to the PD pathological processes.
Collapse
Affiliation(s)
- Antoine Marchand
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Alessia Sarchione
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Panagiotis S. Athanasopoulos
- Department of Cell Biochemistry, University of Groningen, 9747 AG Groningen, The Netherlands; (P.S.A.); (F.Y.H.); (A.K.)
| | | | - Liesel Goveas
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Romain Magnez
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-UMR1277—Canther—Cancer Heterogeneity Plasticity and Resistance to Therapies, Platform of Integrative Chemical Biology, F-59000 Lille, France; (R.M.); (X.T.)
| | - Matthieu Drouyer
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Marco Emanuele
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Franz Y. Ho
- Department of Cell Biochemistry, University of Groningen, 9747 AG Groningen, The Netherlands; (P.S.A.); (F.Y.H.); (A.K.)
| | - Maxime Liberelle
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Patricia Melnyk
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Nicolas Lebègue
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-UMR1277—Canther—Cancer Heterogeneity Plasticity and Resistance to Therapies, Platform of Integrative Chemical Biology, F-59000 Lille, France; (R.M.); (X.T.)
| | - R. Jeremy Nichols
- Department of Pathology, Stanford University, Stanford, CA 94305, USA;
| | - Elisa Greggio
- Physiology, Genetics and Behavior Unit, Department of Biology, University of Padova, 35131 Padova, Italy;
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, 9747 AG Groningen, The Netherlands; (P.S.A.); (F.Y.H.); (A.K.)
| | - Thierry Galli
- Institute of Psychiatry and Neuroscience of Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France;
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | - Marie-Christine Chartier-Harlin
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
- Correspondence: (M.-C.C.-H.); (J.-M.T.)
| | - Jean-Marc Taymans
- University of Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (A.M.); (A.S.); (L.G.); (M.D.); (M.E.); (M.L.); (P.M.); (N.L.)
- Correspondence: (M.-C.C.-H.); (J.-M.T.)
| |
Collapse
|
4
|
Identification of peptides interfering with the LRRK2/PP1 interaction. PLoS One 2020; 15:e0237110. [PMID: 32790695 PMCID: PMC7425875 DOI: 10.1371/journal.pone.0237110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/20/2020] [Indexed: 01/17/2023] Open
Abstract
Serine/threonine phosphatases are responsible for modulating the activities of the protein kinases implicated in the development of several pathologies. Here we identified by a PEP-scan approach a peptide of LRRK2, a Parkinson's disease associated protein, interacting with the phosphatase PP1. In order to study its biological activity, the peptide was fused via its N-terminal to an optimized cell penetrating peptide. We synthesized from the original peptide five interfering peptides and identified two (Mut3DPT-LRRK2-Short and Mut3DPT-LRRK2-Long) able to disrupt the LRRK2/PP1 interaction by competition in anti-LRRK2 immunoprecipitates. Using FITC-labelled peptides, we confirmed their internalization into cell lines as well as into primary cells obtained from healthy or ill human donors. We confirmed by ELISA test the association of Mut3DPT-LRRK2-Long peptide to purified PP1 protein. The peptides Mut3DPT-LRRK2-5 to 8 with either N or C-terminal deletions were not able to disrupt the association LRRK2/PP1 nor to associate with purified PP1 protein. The interfering sequences blocking the PP1/LRRK2 interaction were also fused to a shuttle peptide able to cross the blood brain barrier and showed that the newly generated peptides BBB-LRRK2-Short and BBB-LRRK2-Long were highly resistant to protease degradation. Furthermore, they blocked PP1/LRRK2 interaction and they penetrated into cells. Hence, these newly generated peptides can be employed as new tools in the investigation of the role of the LRRK2/PP1 interaction in normal and pathological conditions.
Collapse
|
5
|
Marchand A, Drouyer M, Sarchione A, Chartier-Harlin MC, Taymans JM. LRRK2 Phosphorylation, More Than an Epiphenomenon. Front Neurosci 2020; 14:527. [PMID: 32612495 PMCID: PMC7308437 DOI: 10.3389/fnins.2020.00527] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in the Leucine Rich Repeat Kinase 2 (LRRK2) gene are linked to autosomal dominant Parkinson's disease (PD), and genetic variations at the LRRK2 locus are associated with an increased risk for sporadic PD. This gene encodes a kinase that is physiologically multiphosphorylated, including clusters of both heterologous phosphorylation and autophosphorylation sites. Several pieces of evidence indicate that LRRK2's phosphorylation is important for its pathological and physiological functioning. These include a reduced LRRK2 heterologous phosphorylation in PD brains or after pharmacological inhibition of LRRK2 kinase activity as well as the appearance of subcellular LRRK2 accumulations when this protein is dephosphorylated at heterologous phosphosites. Nevertheless, the regulatory mechanisms governing LRRK2 phosphorylation levels and the cellular consequences of changes in LRRK2 phosphorylation remain incompletely understood. In this review, we present current knowledge on LRRK2 phosphorylation, LRRK2 phosphoregulation, and how LRRK2 phosphorylation changes affect cellular processes that may ultimately be linked to PD mechanisms.
Collapse
Affiliation(s)
- Antoine Marchand
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| | - Matthieu Drouyer
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| | - Alessia Sarchione
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| | - Marie-Christine Chartier-Harlin
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| | - Jean-Marc Taymans
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| |
Collapse
|
6
|
Hanan EJ, Liang J, Wang X, Blake RA, Blaquiere N, Staben ST. Monomeric Targeted Protein Degraders. J Med Chem 2020; 63:11330-11361. [DOI: 10.1021/acs.jmedchem.0c00093] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
7
|
Correddu D, Sharma N, Kaur S, Varnava KG, Mbenza NM, Sarojini V, Leung IKH. An investigation into the effect of ribosomal protein S15 phosphorylation on its intermolecular interactions by using phosphomimetic mutant. Chem Commun (Camb) 2020; 56:7857-7860. [DOI: 10.1039/d0cc01618g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An investigation using recombinant ribosomal proteins and synthetic peptide models was conducted to uncover the effect of the introduction of a negative charge at the C-terminal tail of ribosomal protein S15.
Collapse
Affiliation(s)
- Danilo Correddu
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Nabangshu Sharma
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Simranjeet Kaur
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Kyriakos G. Varnava
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Naasson M. Mbenza
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Vijayalekshmi Sarojini
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology
| | - Ivanhoe K. H. Leung
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery
| |
Collapse
|
8
|
Maekawa T, Tsushima H, Kawakami F, Kawashima R, Kodo M, Imai M, Ichikawa T. Leucine-Rich Repeat Kinase 2 Is Associated With Activation of the Paraventricular Nucleus of the Hypothalamus and Stress-Related Gastrointestinal Dysmotility. Front Neurosci 2019; 13:905. [PMID: 31555076 PMCID: PMC6727664 DOI: 10.3389/fnins.2019.00905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/13/2019] [Indexed: 01/27/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a molecule associated with familial and sporadic Parkinson's disease. It regulates many central neuronal functions, such as cell proliferation, apoptosis, autophagy, and axonal extension. Recently, it has been revealed that LRRK2 is related to anxiety/depression-like behavior, implying an association between LRRK2 and stress. In the present study, we investigated for the first time the stress pathway and its relationship to gastrointestinal motility in LRRK2-knockout (KO) mice. The mice were subjected to acute restraint stress, and analyzed for activation of the paraventricular nucleus of the hypothalamus (PVN) using an immunohistochemical approach. Phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) was assessed by Western blotting. The KO mice showed a lower number of c-Fos-positive cells and disruption of the ERK signaling pathway in the PVN in the presence of restraint stress. Stress responses in terms of both upper and lower gastrointestinal motility were alleviated in the mice, accompanied by lower c-Fos immunoreactivity in enteric excitatory neurons. Our present findings suggest that LRRK2 is a newly recognized molecule regulating the stress pathway in the PVN, playing a role in stress-related gastrointestinal dysmotility.
Collapse
Affiliation(s)
- Tatsunori Maekawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Hiromichi Tsushima
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Behavioral Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Fumitaka Kawakami
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Rei Kawashima
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Masaru Kodo
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Motoki Imai
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| |
Collapse
|
9
|
De Wit T, Baekelandt V, Lobbestael E. Inhibition of LRRK2 or Casein Kinase 1 Results in LRRK2 Protein Destabilization. Mol Neurobiol 2018; 56:5273-5286. [PMID: 30592011 PMCID: PMC6657425 DOI: 10.1007/s12035-018-1449-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/06/2018] [Indexed: 11/25/2022]
Abstract
Mutations and variations in the leucine-rich repeat kinase 2 (LRRK2) gene are strongly associated with an increased risk to develop Parkinson's disease (PD). Most pathogenic LRRK2 mutations display increased kinase activity, which is believed to underlie LRRK2-mediated toxicity. Therefore, major efforts have been invested in the development of potent and selective LRRK2 kinase inhibitors. Several of these compounds have proven beneficial in cells and in vivo, even in a LRRK2 wild-type background. Therefore, LRRK2 kinase inhibition holds great promise as disease-modifying PD therapy, and is currently tested in preclinical and early clinical studies. One of the safety concerns is the development of lung pathology in mice and non-human primates, which is most likely related to the strongly reduced LRRK2 protein levels after LRRK2 kinase inhibition. In this study, we aimed to better understand the molecular consequences of chronic LRRK2 kinase inhibition, which may be pivotal in the further development of a LRRK2 kinase inhibitor-based PD therapy. We found that LRRK2 protein levels are not restored during long-term LRRK2 kinase inhibition, but are recovered upon inhibitor withdrawal. Interestingly, LRRK2 kinase inhibitor-induced destabilization does not occur in all pathogenic LRRK2 variants and the N-terminal part of LRRK2 appears to play a crucial role in this process. In addition, we identified CK1, an upstream kinase of LRRK2, as a regulator of LRRK2 protein stability in cell culture and in vivo. We propose that pharmacological LRRK2 kinase inhibition triggers a cascade that results in reduced CK1-mediated phosphorylation of yet unidentified LRRK2 phosphorylation sites. This process involves the N-terminus of LRRK2 and ultimately leads to LRRK2 protein degradation.
Collapse
Affiliation(s)
- T De Wit
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Herestraat 49 - Bus 1023, 3000, Leuven, Belgium
| | - V Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Herestraat 49 - Bus 1023, 3000, Leuven, Belgium.
| | - E Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Herestraat 49 - Bus 1023, 3000, Leuven, Belgium.
| |
Collapse
|
10
|
Roco Proteins and the Parkinson's Disease-Associated LRRK2. Int J Mol Sci 2018; 19:ijms19124074. [PMID: 30562929 PMCID: PMC6320773 DOI: 10.3390/ijms19124074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 02/08/2023] Open
Abstract
Small G-proteins are structurally-conserved modules that function as molecular on-off switches. They function in many different cellular processes with differential specificity determined by the unique effector-binding surfaces, which undergo conformational changes during the switching action. These switches are typically standalone monomeric modules that form transient heterodimers with specific effector proteins in the 'on' state, and cycle to back to the monomeric conformation in the 'off' state. A new class of small G-proteins called "Roco" was discovered about a decade ago; this class is distinct from the typical G-proteins in several intriguing ways. Their switch module resides within a polypeptide chain of a large multi-domain protein, always adjacent to a unique domain called COR, and its effector kinase often resides within the same polypeptide. As such, the mechanisms of action of the Roco G-proteins are likely to differ from those of the typical G-proteins. Understanding these mechanisms is important because aberrant activity in the human Roco protein LRRK2 is associated with the pathogenesis of Parkinson's disease. This review provides an update on the current state of our understanding of the Roco G-proteins and the prospects of targeting them for therapeutic purposes.
Collapse
|
11
|
Araki M, Ito G, Tomita T. Physiological and pathological functions of LRRK2: implications from substrate proteins. Neuronal Signal 2018; 2:NS20180005. [PMID: 32714591 PMCID: PMC7373236 DOI: 10.1042/ns20180005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) encodes a 2527-amino acid (aa) protein composed of multiple functional domains, including a Ras of complex proteins (ROC)-type GTP-binding domain, a carboxyl terminal of ROC (COR) domain, a serine/threonine protein kinase domain, and several repeat domains. LRRK2 is genetically involved in the pathogenesis of both sporadic and familial Parkinson's disease (FPD). Parkinson's disease (PD) is the second most common neurodegenerative disorder, manifesting progressive motor dysfunction. PD is pathologically characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, and the presence of intracellular inclusion bodies called Lewy bodies (LB) in the remaining neurons. As the most frequent PD-causing mutation in LRRK2, G2019S, increases the kinase activity of LRRK2, an abnormal increase in LRRK2 kinase activity is believed to contribute to PD pathology; however, the precise biological functions of LRRK2 involved in PD pathogenesis remain unknown. Although biochemical studies have discovered several substrate proteins of LRRK2 including Rab GTPases and tau, little is known about whether excess phosphorylation of these substrates is the cause of the neurodegeneration in PD. In this review, we summarize latest findings regarding the physiological and pathological functions of LRRK2, and discuss the possible molecular mechanisms of neurodegeneration caused by LRRK2 and its substrates.
Collapse
Affiliation(s)
- Miho Araki
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Genta Ito
- Laboratory of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
12
|
Abstract
Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are known today as the most common genetic cause of Parkinson's disease (PD). LRRK2 is a large protein that is hypothesized to regulate other proteins as a scaffold in downstream signaling pathways. This is supported by the multiple domain composition of LRRK2 with several protein-protein interaction domains combined with kinase and GTPase activity. LRRK2 is highly phosphorylated at sites that are strictly controlled by upstream regulators, including its own kinase domain. In cultured cells, most pathogenic mutants display increased autophosphorylation at S1292, but decreased phosphorylation at sites controlled by other kinases. We only begin to understand how LRRK2 phosphorylation is regulated and how this impacts its physiological and pathological function. Intriguingly, LRRK2 kinase inhibition, currently one of the most prevailing disease-modifying therapeutic strategies for PD, induces LRRK2 dephosphorylation at sites that are also dephosphorylated in pathogenic variants. In addition, LRRK2 kinase inhibition can induce LRRK2 protein degradation, which might be related to the observed inhibitor-induced adverse effects on the lung in rodents and non-human primates, as it resembles the lung pathology in LRRK2 knock-out animals. In this review, we will provide an overview of how LRRK2 phosphorylation is regulated and how this complex regulation relates to several molecular and cellular features of LRRK2.
Collapse
Affiliation(s)
- Tina De Wit
- 1 Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- 1 Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Evy Lobbestael
- 1 Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Chen Z, Cao Z, Zhang W, Gu M, Zhou ZD, Li B, Li J, Tan EK, Zeng L. LRRK2 interacts with ATM and regulates Mdm2–p53 cell proliferation axis in response to genotoxic stress. Hum Mol Genet 2017; 26:4494-4505. [DOI: 10.1093/hmg/ddx337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/06/2017] [Indexed: 01/29/2023] Open
|
14
|
LRRK2 detection in human biofluids: potential use as a Parkinson's disease biomarker? Biochem Soc Trans 2017; 45:207-212. [PMID: 28202674 DOI: 10.1042/bst20160334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 02/05/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a complex signalling protein that is a key therapeutic target, particularly in Parkinson's disease (PD). In addition, there is now evidence showing that LRRK2 expression and phosphorylation levels have potential as markers of disease or target engagement. Indeed, reports show increases in LRRK2 protein levels in the prefrontal cortex of PD patients relative to controls, suggesting that increase in total LRRK2 protein expression is correlated with disease progression. LRRK2 phosphorylation levels are reduced in experimental systems for most disease mutants, and LRRK2 is also rapidly dephosphorylated upon LRRK2 inhibitor treatment, considered potential therapeutics. Recently, the presence of LRRK2 was confirmed in exosomes from human biofluids, including urine and cerebrospinal fluid. Moreover, phosphorylation of LRRK2 at phosphosites S910, S935, S955 and S973, as well as at the autophosphoryation site S1292, was found in urinary exosomes. In this review, we summarize knowledge on detection of LRRK2 in human biofluids and the relevance of these findings for the development of PD-related biomarkers.
Collapse
|
15
|
Abstract
LRRK2 is a highly phosphorylated protein, and evidence of a physiological role for LRRK2 phosphorylation has accumulated in recent years for cellular phosphosites, many of which are found in the ANK-LRR interdomain region, i.e., the S910/S935/S955/S973 sites as well as recently for autophosphorylation sites, at least one of which has been confirmed in cells, S1292. LRRK2 phosphorylation is modulated in several disease or potential therapy relevant conditions such as in disease mutant variants of LRRK2 or following LRRK2 kinase inhibitor treatment. This chapter will focus on the regulation of LRRK2 phosphorylation and more specifically the role of phosphatases in LRRK2 dephosphorylation. This will include reviewing the conditions in which LRRK2 is found to be dephosphorylated, the molecular partners and phosphatases involved in regulating LRRK2 phosphorylation, as well as discussing how LRRK2 phosphatases may be therapeutic targets or biomarkers in their own right.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Université de Lille, Inserm, CHU Lille, UMR-S 1172-JPARC Jean-Pierre Aubert Research Center, Neurosciences and Cancer, 1 Place de Verdun, Lille, 59000, France.
| |
Collapse
|
16
|
Ding X, Barodia SK, Ma L, Goldberg MS. Fbxl18 targets LRRK2 for proteasomal degradation and attenuates cell toxicity. Neurobiol Dis 2016; 98:122-136. [PMID: 27890708 DOI: 10.1016/j.nbd.2016.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 11/09/2016] [Accepted: 11/17/2016] [Indexed: 01/18/2023] Open
Abstract
Dominantly inherited mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common causes of familial Parkinson's disease (PD) and LRRK2 polymorphisms are associated with increased risk for idiopathic PD. However, the molecular mechanisms by which these mutations cause PD remain uncertain. In vitro studies indicate that disease-linked mutations in LRRK2 increase LRRK2 kinase activity and LRRK2-mediated cell toxicity. Identifying LRRK2-interacting proteins and determining their effects on LRRK2 are important for understanding LRRK2 function and for delineating the pathophysiological mechanisms of LRRK2 mutations. Here we identified a novel protein, F-box and leucine-rich repeat domain-containing protein 18 (Fbxl18) that physically associates with LRRK2. We demonstrated that Fbxl18 is a component of a Skp1-Cullin1-F-box ubiquitin ligase complex that regulates the abundance of LRRK2 by selectively targeting phosphorylated LRRK2 for ubiquitination and proteasomal degradation. Knockdown of endogenous Fbxl18 stabilized LRRK2 abundance while protein kinase C activation enhanced LRRK2 degradation by Fbxl18. Dephosphorylation of LRRK2 blocked Fbxl18 association with LRRK2. Taken together, we have identified potential mechanisms for LRRK2 regulation by kinase signaling pathways. Furthermore, Fbxl18 prevented caspase activation and cell death caused by LRRK2 and PD-linked mutant LRRK2. This reveals novel targets for developing potential therapies for familial and idiopathic PD.
Collapse
Affiliation(s)
- Xiaodong Ding
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sandeep K Barodia
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lisha Ma
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew S Goldberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
Lobbestael E, Civiero L, De Wit T, Taymans JM, Greggio E, Baekelandt V. Pharmacological LRRK2 kinase inhibition induces LRRK2 protein destabilization and proteasomal degradation. Sci Rep 2016; 6:33897. [PMID: 27658356 PMCID: PMC5034242 DOI: 10.1038/srep33897] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/06/2016] [Indexed: 02/07/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) kinase activity is increased in several pathogenic mutations, including the most common mutation, G2019S, and is known to play a role in Parkinson’s disease (PD) pathobiology. This has stimulated the development of potent, selective LRRK2 kinase inhibitors as one of the most prevailing disease-modifying therapeutic PD strategies. Although several lines of evidence support beneficial effects of LRRK2 kinase inhibitors, many questions need to be answered before clinical applications can be envisaged. Using six different LRRK2 kinase inhibitors, we show that LRRK2 kinase inhibition induces LRRK2 dephosphorylation and can reduce LRRK2 protein levels of overexpressed wild type and G2019S, but not A2016T or K1906M, LRRK2 as well as endogenous LRRK2 in mouse brain, lung and kidney. The inhibitor-induced reduction in LRRK2 levels could be reversed by proteasomal inhibition, but not by lysosomal inhibition, while mRNA levels remained unaffected. In addition, using LRRK2 S910A and S935A phosphorylation mutants, we show that dephosphorylation of these sites is not required for LRRK2 degradation. Increasing our insight in the molecular and cellular consequences of LRRK2 kinase inhibition will be crucial in the further development of LRRK2-based PD therapies.
Collapse
Affiliation(s)
- E Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium
| | - L Civiero
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - T De Wit
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium
| | - J-M Taymans
- UMR-S1172 Jean-Pierre Aubert Research Center - (INSERM - CHRU de Lille - Université de Lille), Early Stages of Parkinson's Disease Team, Lille, France
| | - E Greggio
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - V Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Kapucijnenvoer 33, 3000 Leuven, Belgium
| |
Collapse
|
18
|
Athanasopoulos PS, Jacob W, Neumann S, Kutsch M, Wolters D, Tan EK, Bichler Z, Herrmann C, Heumann R. Identification of protein phosphatase 2A as an interacting protein of leucine-rich repeat kinase 2. Biol Chem 2016; 397:541-54. [DOI: 10.1515/hsz-2015-0189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 02/17/2016] [Indexed: 11/15/2022]
Abstract
Abstract
Mutations in the gene coding for the multi-domain protein leucine-rich repeat kinase 2 (LRRK2) are the leading cause of genetically inherited Parkinson’s disease (PD). Two of the common found mutations are the R1441C and G2019S. In this study we identified protein phosphatase 2A (PP2A) as an interacting partner of LRRK2. We were able to demonstrate that the Ras of complex protein (ROC) domain is sufficient to interact with the three subunits of PP2A in human neuroblastoma SH-SY5Y cells and in HeLa cells. The alpha subunit of PP2A is interacting with LRRK2 in the perinuclear region of HeLa cells. Silencing the catalytic subunit of PP2A by shRNA aggravated cellular degeneration induced by the pathogenic R1441C-LRRK2 mutant expressed in neuroblastoma SH-SY5Y cells. A similar enhancement of apoptotic nuclei was observed by downregulation of the catalytic subunit of PP2A in cultured cortical cells derived from neurons overexpressing the pathogenic mutant G2019S-LRRK2. Conversely, pharmacological activation of PP2A by sodium selenate showed a partial neuroprotection from R1441C-LRRK2-induced cellular degeneration. All these data suggest that PP2A is a new interacting partner of LRRK2 and reveal the importance of PP2A as a potential therapeutic target in PD.
Collapse
|
19
|
Taymans JM, Greggio E. LRRK2 Kinase Inhibition as a Therapeutic Strategy for Parkinson's Disease, Where Do We Stand? Curr Neuropharmacol 2016; 14:214-25. [PMID: 26517051 PMCID: PMC4857626 DOI: 10.2174/1570159x13666151030102847] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022] Open
Abstract
One of the most promising therapeutic targets for potential disease-modifying treatment of Parkinson's disease (PD) is leucine-rich repeat kinase 2 (LRRK2). Specifically, targeting LRRK2's kinase function has generated a lot of interest from both industry and academia. This work has yielded several published studies showing the feasibility of developing potent, selective and brain permeable LRRK2 kinase inhibitors. The availability of these experimental drugs is contributing to filling in the gaps in our knowledge on the safety and efficacy of LRRK2 kinase inhibition. Recent studies of LRRK2 kinase inhibition in preclinical models point to potential undesired effects in peripheral tissues such as lung and kidney. Also, while strategies are now emerging to measure target engagement of LRRK2 inhibitors, there remains an important need to expand efficacy studies in preclinical models of progressive PD. Future work in the LRRK2 inhibition field must therefore be directed towards developing molecules and treatment regimens which demonstrate efficacy in mammalian models of disease in conditions where safety liabilities are reduced to a minimum.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Jean-Pierre Aubert Research Center, UMR-S1172,rue Polonovski - 1 place de Verdun, 59045 Lille, France.
| | - Elisa Greggio
- Department of Biology, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
20
|
Ho DH, Kim H, Kim J, Sim H, Ahn H, Kim J, Seo H, Chung KC, Park BJ, Son I, Seol W. Leucine-Rich Repeat Kinase 2 (LRRK2) phosphorylates p53 and induces p21(WAF1/CIP1) expression. Mol Brain 2015; 8:54. [PMID: 26384650 PMCID: PMC4575451 DOI: 10.1186/s13041-015-0145-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/04/2015] [Indexed: 11/25/2022] Open
Abstract
Background Leucine-rich repeat kinase 2 (LRRK2) is a gene in which a mutation causes Parkinson’s disease (PD), and p53 is a prototype tumor suppressor. In addition, activation of p53 in patient with PD has been reported by several studies. Because phosphorylation of p53 is critical for regulating its activity and LRRK2 is a kinase, we tested whether p53 is phosphorylated by LRRK2. Results LRRK2 phosphorylates threonine (Thr) at TXR sites in an in vitro kinase assay, and the T304 and T377 were identified as putative phosphorylated residues. An increase of phospho-Thr in the p53 TXR motif was confirmed in the cells overexpressing G2019S, and human induced pluripotent stem (iPS) cells of a G2019S carrier. Interactions between LRRK2 and p53 were confirmed by co-immunoprecipitation of lysates of differentiated SH-SY5Y cells. LRRK2 mediated p53 phosphorylation translocalizes p53 predominantly to nucleus and increases p21WAF1/CIP1 expression in SH-SY5Y cells based on reverse transcription-polymerase chain reaction and Western blot assay results. The luciferase assay using the p21WAF1/CIP1 promoter-reporter also confirmed that LRRK2 kinase activity increases p21 expression. Exogenous expression of G2019S and the phosphomimetic p53 T304/377D mutants increased expression of p21WAF1/CIP1 and cleaved PARP, and cytotoxicity in the same cells. We also observed increase of p21 expression in rat primary neuron cells after transient expression of p53 T304/377D mutants and the mid-brain lysates of the G2019S transgenic mice. Conclusion p53 is a LRRK2 kinase substrate. Phosphorylation of p53 by LRRK2 induces p21WAF1/CIP1 expression and apoptosis in differentiated SH-SY5Y cells and rat primary neurons. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0145-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.,Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyejung Kim
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea
| | - Jisun Kim
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyuna Sim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyunjun Ahn
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seodaemun-gu, Seoul, Republic of Korea
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan, Republic of Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea. .,Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| |
Collapse
|
21
|
Ohta E, Nihira T, Uchino A, Imaizumi Y, Okada Y, Akamatsu W, Takahashi K, Hayakawa H, Nagai M, Ohyama M, Ryo M, Ogino M, Murayama S, Takashima A, Nishiyama K, Mizuno Y, Mochizuki H, Obata F, Okano H. I2020T mutant LRRK2 iPSC-derived neurons in the Sagamihara family exhibit increased Tau phosphorylation through the AKT/GSK-3β signaling pathway. Hum Mol Genet 2015; 24:4879-900. [PMID: 26056228 DOI: 10.1093/hmg/ddv212] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/03/2015] [Indexed: 12/22/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is the causative molecule of the autosomal dominant hereditary form of Parkinson's disease (PD), PARK8, which was originally defined in a study of a Japanese family (the Sagamihara family) harboring the I2020T mutation in the kinase domain. Although a number of reported studies have focused on cell death mediated by mutant LRRK2, details of the pathogenetic effect of LRRK2 still remain to be elucidated. In the present study, to elucidate the mechanism of neurodegeneration in PD caused by LRRK2, we generated induced pluripotent stem cells (iPSC) derived from fibroblasts of PD patients with I2020T LRRK2 in the Sagamihara family. We found that I2020T mutant LRRK2 iPSC-derived neurons released less dopamine than control-iPSC-derived neurons. Furthermore, we demonstrated that patient iPSC-derived neurons had a lower phospho-AKT level than control-iPSC-derived neurons, and that the former showed an increased incidence of apoptosis relative to the controls. Interestingly, patient iPSC-derived neurons exhibited activation of glycogen synthase kinase-3β (GSK-3β) and high Tau phosphorylation. In addition, the postmortem brain of the patient from whom the iPSC had been established exhibited deposition of neurofibrillary tangles as well as increased Tau phosphorylation in neurons. These results suggest that I2020T LRRK2-iPSC could be a promising new tool for reproducing the pathology of PD in the brain caused by the I2020T mutation, and applicable as a model in studies of targeted therapeutics.
Collapse
Affiliation(s)
- Etsuro Ohta
- R & D Center for Cell Design, Institute for Regenerative Medicine and Cell Design, Kitasato University School of Allied Health Sciences, Kanagawa, Japan, Division of Clinical Immunology, Graduate School of Medical Sciences, Department of Physiology
| | - Tomoko Nihira
- Department of Neuro-Regenerative Medicine, Department of Physiology
| | - Akiko Uchino
- Department of Neurology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan, Department of the Brain Bank for Aging Research
| | | | - Yohei Okada
- Department of Physiology, Department of Neurology, Aichi Medical University School of Medicine, Aichi, Japan
| | - Wado Akamatsu
- Department of Physiology, Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Kayoko Takahashi
- Department of Medical Laboratory, Kitasato University Hospital, Kanagawa, Japan
| | | | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Manabu Ohyama
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masafuchi Ryo
- Department of Neurology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mieko Ogino
- Department of Neurology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Shigeo Murayama
- Department of the Brain Bank for Aging Research, Department of Neurology, Department of Bioresource Center (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Akihiko Takashima
- Department of Neurobiology, National Center for Geriatrics and Gerontology, Obu, Japan and
| | - Kazutoshi Nishiyama
- Department of Neurology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan, Department of Neurology, Kitasato University School of Medicine, Kanagawa, Japan
| | | | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumiya Obata
- R & D Center for Cell Design, Institute for Regenerative Medicine and Cell Design, Kitasato University School of Allied Health Sciences, Kanagawa, Japan, Division of Clinical Immunology, Graduate School of Medical Sciences
| | | |
Collapse
|
22
|
The Role of α-Synuclein and LRRK2 in Tau Phosphorylation. PARKINSONS DISEASE 2015; 2015:734746. [PMID: 25977830 PMCID: PMC4419261 DOI: 10.1155/2015/734746] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 01/08/2023]
Abstract
There is now a considerable body of experimental evidence that Parkinson's disease arises through physiological interaction of causative molecules, leading to tau pathology. In this review, we discuss the physiological role of α-synuclein and LRRK2 in the abnormal phosphorylation of tau. In addition, as recent reports have indicated that heat shock proteins- (HSPs-) inducing drugs can help to ameliorate neurodegenerative diseases associated with tau pathology, we also discuss therapeutic strategies for PD focusing on inhibition of α-synuclein- and LRRK2-associated tau phosphorylation by HSPs.
Collapse
|
23
|
Krumova P, Reyniers L, Meyer M, Lobbestael E, Stauffer D, Gerrits B, Muller L, Hoving S, Kaupmann K, Voshol J, Fabbro D, Bauer A, Rovelli G, Taymans JM, Bouwmeester T, Baekelandt V. Chemical genetic approach identifies microtubule affinity-regulating kinase 1 as a leucine-rich repeat kinase 2 substrate. FASEB J 2015; 29:2980-92. [PMID: 25854701 DOI: 10.1096/fj.14-262329] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/09/2015] [Indexed: 12/25/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of autosomal-dominant forms of Parkinson's disease. LRRK2 is a modular, multidomain protein containing 2 enzymatic domains, including a kinase domain, as well as several protein-protein interaction domains, pointing to a role in cellular signaling. Although enormous efforts have been made, the exact pathophysiologic mechanisms of LRRK2 are still not completely known. In this study, we used a chemical genetics approach to identify LRRK2 substrates from mouse brain. This approach allows the identification of substrates of 1 particular kinase in a complex cellular environment. Several of the identified peptides are involved in the regulation of microtubule (MT) dynamics, including microtubule-associating protein (MAP)/microtubule affinity-regulating kinase 1 (MARK1). MARK1 is a serine/threonine kinase known to phosphorylate MT-binding proteins such as Tau, MAP2, and MAP4 at KXGS motifs leading to MT destabilization. In vitro kinase assays and metabolic-labeling experiments in living cells confirmed MARK1 as an LRRK2 substrate. Moreover, we also showed that LRRK2 and MARK1 are interacting in eukaryotic cells. Our findings contribute to the identification of physiologic LRRK2 substrates and point to a potential mechanism explaining the reported effects of LRRK2 on neurite morphology.
Collapse
Affiliation(s)
- Petranka Krumova
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Lauran Reyniers
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Marc Meyer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Evy Lobbestael
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Daniela Stauffer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Bertran Gerrits
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Lionel Muller
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sjouke Hoving
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Klemens Kaupmann
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Johannes Voshol
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Doriano Fabbro
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Andreas Bauer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Giorgio Rovelli
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Jean-Marc Taymans
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Tewis Bouwmeester
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Porras P, Duesbury M, Fabregat A, Ueffing M, Orchard S, Gloeckner CJ, Hermjakob H. A visual review of the interactome of LRRK2: Using deep-curated molecular interaction data to represent biology. Proteomics 2015; 15:1390-404. [PMID: 25648416 PMCID: PMC4415485 DOI: 10.1002/pmic.201400390] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 01/15/2015] [Accepted: 01/29/2015] [Indexed: 02/04/2023]
Abstract
Molecular interaction databases are essential resources that enable access to a wealth of information on associations between proteins and other biomolecules. Network graphs generated from these data provide an understanding of the relationships between different proteins in the cell, and network analysis has become a widespread tool supporting –omics analysis. Meaningfully representing this information remains far from trivial and different databases strive to provide users with detailed records capturing the experimental details behind each piece of interaction evidence. A targeted curation approach is necessary to transfer published data generated by primarily low-throughput techniques into interaction databases. In this review we present an example highlighting the value of both targeted curation and the subsequent effective visualization of detailed features of manually curated interaction information. We have curated interactions involving LRRK2, a protein of largely unknown function linked to familial forms of Parkinson's disease, and hosted the data in the IntAct database. This LRRK2-specific dataset was then used to produce different visualization examples highlighting different aspects of the data: the level of confidence in the interaction based on orthogonal evidence, those interactions found under close-to-native conditions, and the enzyme–substrate relationships in different in vitro enzymatic assays. Finally, pathway annotation taken from the Reactome database was overlaid on top of interaction networks to bring biological functional context to interaction maps.
Collapse
Affiliation(s)
- Pablo Porras
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Yun HJ, Kim H, Ga I, Oh H, Ho DH, Kim J, Seo H, Son I, Seol W. An early endosome regulator, Rab5b, is an LRRK2 kinase substrate. J Biochem 2015; 157:485-95. [PMID: 25605758 DOI: 10.1093/jb/mvv005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/18/2014] [Indexed: 11/13/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been identified as a causative gene for Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, both of which provide critical intracellular signal-transduction functions. We showed previously that Rab5b, a small GTPase protein that regulates the motility and fusion of early endosomes, interacts with LRRK2 and co-regulates synaptic vesicle endocytosis. Using recombinant proteins, we show here that LRRK2 phosphorylates Rab5b at its Thr6 residue in in vitro kinase assays with mass spectrophotometry analysis. Phosphorylation of Rab5b by LRRK2 on the threonine residue was confirmed by western analysis using cells stably expressing LRRK2 G2019S. The phosphomimetic T6D mutant exhibited stronger GTPase activity than that of the wild-type Rab5b. In addition, phosphorylation of Rab5b by LRRK2 also exhibited GTPase activity stronger than that of the unphosphorylated Rab5b protein. Two assays testing Rab5's activity, neurite outgrowth analysis and epidermal growth factor receptor degradation assays, showed that Rab5b T6D exhibited phenotypes that were expected to be observed in the inactive Rab5b, including longer neurite length and less degradation of EGFR. These results suggest that LRRK2 kinase activity functions as a Rab5b GTPase activating protein and thus, negatively regulates Rab5b signalling.
Collapse
Affiliation(s)
- Hye Jin Yun
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Hyejung Kim
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Inhwa Ga
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Hakjin Oh
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Dong Hwan Ho
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Jiyoung Kim
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Hyemyung Seo
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Ilhong Son
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| | - Wongi Seol
- Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea Institute for Brain Science and Technology, Inje University, Gaegumdong, Busanjingu, Busan, South Korea; InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea; Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, South Korea; and Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbondong, Gunposhi, Gyeonggido, South Korea
| |
Collapse
|
26
|
Phosphorylation of LRRK2 by casein kinase 1α regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat Commun 2014; 5:5827. [PMID: 25500533 PMCID: PMC4268884 DOI: 10.1038/ncomms6827] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/10/2014] [Indexed: 12/29/2022] Open
Abstract
LRRK2, a gene relevant to Parkinson's disease, encodes a scaffolding protein with both GTPase and kinase activities. LRRK2 protein is itself phosphorylated and therefore subject to regulation by cell signaling but the kinase(s) responsible for this event have not been definitively identified. Here, using an unbiased siRNA kinome screen, we identify and validate casein kinase 1α (CK1α) as being responsible for LRRK2 phosphorylation, including in the adult mouse striatum. We further show that LRRK2 recruitment to TGN46-positive Golgi-derived vesicles is modulated by constitutive LRRK2 phosphorylation by CK1α. These effects are mediated by differential protein interactions of LRRK2 with a guanine nucleotide exchange factor, ARHGEF7. These pathways are therefore likely involved in the physiological maintenance of the Golgi in cells, which may play a role in the pathogenesis of Parkinson's disease.
Collapse
|
27
|
Taymans JM, Baekelandt V. Phosphatases of α-synuclein, LRRK2, and tau: important players in the phosphorylation-dependent pathology of Parkinsonism. Front Genet 2014; 5:382. [PMID: 25426138 PMCID: PMC4224088 DOI: 10.3389/fgene.2014.00382] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
An important challenge in the field of Parkinson’s disease (PD) is to develop disease modifying therapies capable of stalling or even halting disease progression. Coupled to this challenge is the need to identify disease biomarkers, in order to identify pre-symptomatic hallmarks of disease and monitor disease progression. The answer to these challenges lies in the elucidation of the molecular causes underlying PD, for which important leads are disease genes identified in studies investigating the underlying genetic causes of PD. LRRK2 and α-syn have been both linked to familial forms of PD as well as associated to sporadic PD. Another gene, microtubule associated protein tau (MAPT), has been genetically linked to a dominant form of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and genome-wide association studies report a strong association between MAPT and sporadic PD. Interestingly, LRRK2, α-syn, and tau are all phosphorylated proteins, and their phosphorylation patterns are linked to disease. In this review, we provide an overview of the evidence linking LRRK2, α-syn, and tau phosphorylation to PD pathology and focus on studies which have identified phosphatases responsible for dephosphorylation of pathology-related phosphorylations. We also discuss how the LRRK2, α-syn, and tau phosphatases may point to separate or cross-talking pathological pathways in PD. Finally, we will discuss how the study of phosphatases of dominant Parkinsonism proteins opens perspectives for targeting pathological phosphorylation events.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| |
Collapse
|
28
|
Taymans JM, Baekelandt V, Harvey K. Regulation and targeting of enzymes mediating Parkinson's disease pathogenesis: focus on Parkinson's disease kinases, GTPases, and ATPases. Front Mol Neurosci 2014; 7:71. [PMID: 25120428 PMCID: PMC4114254 DOI: 10.3389/fnmol.2014.00071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/09/2014] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences KU Leuven, Leuven, Belgium
| | - Kirsten Harvey
- School of Pharmacy, University College London London, UK
| |
Collapse
|
29
|
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. [PMID: 24917786 PMCID: PMC4042160 DOI: 10.3389/fnmol.2014.00051] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/14/2014] [Indexed: 01/23/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a complex, multidomain protein which is considered a valuable target for potential disease-modifying therapeutic strategies for Parkinson's disease (PD). In mammalian cells and brain, LRRK2 is phosphorylated and treatment of cells with inhibitors of LRRK2 kinase activity can induce LRRK2 dephosphorylation at a cluster of serines including Ser910/935/955/973. It has been suggested that phosphorylation levels at these sites reflect LRRK2 kinase activity, however kinase-dead variants of LRRK2 or kinase activating variants do not display altered Ser935 phosphorylation levels compared to wild type. Furthermore, Ser910/935/955/973 are not autophosphorylation sites, therefore, it is unclear if inhibitor induced dephosphorylation depends on the activity of compounds on LRRK2 or on yet to be identified upstream kinases. Here we used a panel of 160 ATP competitive and cell permeable kinase inhibitors directed against all branches of the kinome and tested their activity on LRRK2 in vitro using a peptide-substrate-based kinase assay. In neuronal SH-SY5Y cells overexpressing LRRK2 we used compound-induced dephosphorylation of Ser935 as readout. In silico docking of selected compounds was performed using a modeled LRRK2 kinase structure. Receiver operating characteristic plots demonstrated that the obtained docking scores to the LRRK2 ATP binding site correlated with in vitro and cellular compound activity. We also found that in vitro potency showed a high degree of correlation to cellular compound induced LRRK2 dephosphorylation activity across multiple compound classes. Therefore, acute LRRK2 dephosphorylation at Ser935 in inhibitor treated cells involves a strong component of inhibitor activity on LRRK2 itself, without excluding a role for upstream kinases. Understanding the regulation of LRRK2 phosphorylation by kinase inhibitors aids our understanding of LRRK2 signaling and may lead to development of new classes of LRRK2 kinase inhibitors.
Collapse
Affiliation(s)
- Renée Vancraenenbroeck
- Laboratory for Biomolecular Modelling, Division of Biochemistry, Molecular and Structural Biology, Department of Chemistry, KU Leuven Leuven, Belgium
| | - Joren De Raeymaecker
- Laboratory for Biomolecular Modelling, Division of Biochemistry, Molecular and Structural Biology, Department of Chemistry, KU Leuven Leuven, Belgium
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Fangye Gao
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Marc De Maeyer
- Laboratory for Biomolecular Modelling, Division of Biochemistry, Molecular and Structural Biology, Department of Chemistry, KU Leuven Leuven, Belgium
| | - Arnout Voet
- Laboratory for Biomolecular Modelling, Division of Biochemistry, Molecular and Structural Biology, Department of Chemistry, KU Leuven Leuven, Belgium ; Zhang Initiative Research Unit, Riken Saitama, Japan
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| |
Collapse
|
30
|
Lack of correlation between the kinase activity of LRRK2 harboring kinase-modifying mutations and its phosphorylation at Ser910, 935, and Ser955. PLoS One 2014; 9:e97988. [PMID: 24836358 PMCID: PMC4024040 DOI: 10.1371/journal.pone.0097988] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 04/27/2014] [Indexed: 01/01/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is extensively phosphorylated in cells within a region amino-terminal to the leucine-rich repeat domain. Since phosphorylation in this region of LRRK2, including Ser910, Ser935, Ser955, and Ser973, is significantly downregulated upon treatment with inhibitors of LRRK2, it has been hypothesized that signaling pathways downstream of the kinase activity of LRRK2 are involved in regulating the phosphorylation of LRRK2, although the precise mechanism has remained unknown. Here we examined the effects of LRRK2 inhibitors on the phosphorylation state at Ser910, Ser935, and Ser955 in a series of kinase-inactive mutants of LRRK2. We found that the responses of LRRK2 to the inhibitors varied among mutants, in a manner not consistent with the above-mentioned hypothesis. Notably, one of the kinase-inactive mutants, T2035A LRRK2, underwent phosphorylation, as well as the inhibitor-induced dephosphorylation, at Ser910, Ser935, and Ser955, to a similar extent to those observed with wild-type LRRK2. These results suggest that the kinase activity of LRRK2 is not involved in the common mechanism of inhibitor-induced dephosphorylation of LRRK2.
Collapse
|
31
|
Gilsbach BK, Kortholt A. Structural biology of the LRRK2 GTPase and kinase domains: implications for regulation. Front Mol Neurosci 2014; 7:32. [PMID: 24847205 PMCID: PMC4017136 DOI: 10.3389/fnmol.2014.00032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022] Open
Abstract
Human leucine rich repeat kinase 2 (LRRK2) belongs to the Roco family of proteins, which are characterized by the presence of a Ras-like G-domain (Roc), a C-terminal of Roc domain (COR), and a kinase domain. Mutations in LRRK2 have been found to be thus far the most frequent cause of late-onset Parkinson’s disease (PD). Several of the pathogenic mutations in LRRK2 result in decreased GTPase activity and enhanced kinase activity, suggesting a possible PD-related gain of abnormal function. Important progress in the structural understanding of LRRK2 has come from our work with related Roco proteins from lower organisms. Atomic structures of Roco proteins from prokaryotes revealed that Roco proteins belong to the GAD class of molecular switches (G proteins activated by nucleotide dependent dimerization). As in LRRK2, PD-analogous mutations in Roco proteins from bacteria decrease the GTPase reaction. Studies with Roco proteins from the model organism Dictyostelium discoideum revealed that PD mutants have different effects and most importantly they explained the G2019S-related increased LRRK2 kinase activity. Furthermore, the structure of Dictyostelium Roco4 kinase in complex with the LRRK2 inhibitor H1152 showed that Roco4 and other Roco family proteins can be important for the optimization of the current, and identification of new, LRRK2 kinase inhibitors. In this review we highlight the recent progress in structural and biochemical characterization of Roco proteins and discuss its implication for the understanding of the complex regulatory mechanism of LRRK2.
Collapse
Affiliation(s)
- Bernd K Gilsbach
- Department of Cell Biochemistry, University of Groningen Groningen, Netherlands
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen Groningen, Netherlands
| |
Collapse
|
32
|
Silva RG, Geoghegan KF, Qiu X, Aulabaugh A. A continuous and direct assay to monitor leucine-rich repeat kinase 2 activity. Anal Biochem 2014; 450:63-9. [DOI: 10.1016/j.ab.2014.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 12/01/2022]
|
33
|
Mills RD, Mulhern TD, Liu F, Culvenor JG, Cheng HC. Prediction of the Repeat Domain Structures and Impact of Parkinsonism-Associated Variations on Structure and Function of all Functional Domains of Leucine-Rich Repeat Kinase 2 (LRRK2). Hum Mutat 2014; 35:395-412. [DOI: 10.1002/humu.22515] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 01/08/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Ryan D. Mills
- Department of Biochemistry and Molecular Biology; Bio21 Molecular Science and Biotechnology Institute; University of Melbourne; Parkville Victoria Australia
| | - Terrence D. Mulhern
- Department of Biochemistry and Molecular Biology; Bio21 Molecular Science and Biotechnology Institute; University of Melbourne; Parkville Victoria Australia
| | - Fei Liu
- Department of Chemistry & Biomolecular Sciences; Macquarie University; NSW Australia
| | - Janetta G. Culvenor
- Department of Pathology; University of Melbourne; Parkville Victoria Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology; Bio21 Molecular Science and Biotechnology Institute; University of Melbourne; Parkville Victoria Australia
| |
Collapse
|
34
|
Identification of protein phosphatase 1 as a regulator of the LRRK2 phosphorylation cycle. Biochem J 2013; 456:119-28. [PMID: 23937259 DOI: 10.1042/bj20121772] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A cluster of phosphorylation sites in LRRK2 (leucine-rich repeat kinase 2), including Ser910, Ser935, Ser955 and Ser973, is important for PD (Parkinson's disease) pathogenesis as several PD-linked LRRK2 mutants are dephosphorylated at these sites. LRRK2 is also dephosphorylated in cells after pharmacological inhibition of its kinase activity, which is currently proposed as a strategy for disease-modifying PD therapy. Despite this importance of LRRK2 dephosphorylation in mutant LRRK2 pathological mechanism(s) and in LRRK2's response to inhibition, the mechanism by which this occurs is unknown. Therefore we aimed to identify the phosphatase for LRRK2. Using a panel of recombinant phosphatases, we found that PP1 (protein phosphatase 1) efficiently dephosphorylates LRRK2 in vitro. PP1 activity on LRRK2 dephosphorylation was confirmed in cells using PP1 inhibition to reverse LRRK2 dephosphorylation induced by the potent LRRK2 kinase inhibitor LRRK2-IN1 as well as in R1441G mutant LRRK2. We also found that PP1 and LRRK2 can form a complex in cells. Furthermore, we observed that PP1 inhibition modulates LRRK2's cellular phenotype by reducing skein-like LRRK2-positive structures associated with dephosphorylation. In conclusion, the present study reveals PP1 as the physiological LRRK2 phosphatase, responsible for LRRK2 dephosphorylation observed in PD mutant LRRK2 and after LRRK2 kinase inhibition.
Collapse
|
35
|
Parkinson-related LRRK2 mutation R1441C/G/H impairs PKA phosphorylation of LRRK2 and disrupts its interaction with 14-3-3. Proc Natl Acad Sci U S A 2013; 111:E34-43. [PMID: 24351927 DOI: 10.1073/pnas.1312701111] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a multidomain protein implicated in Parkinson disease (PD); however, the molecular mechanism and mode of action of this protein remain elusive. cAMP-dependent protein kinase (PKA), along with other kinases, has been suggested to be an upstream kinase regulating LRRK2 function. Using MS, we detected several sites phosphorylated by PKA, including phosphorylation sites within the Ras of complex proteins (ROC) GTPase domain as well as some previously described sites (S910 and S935). We systematically mapped those sites within LRRK2 and investigated their functional consequences. S1444 in the ROC domain was confirmed as a target for PKA phosphorylation using ROC single-domain constructs and through site-directed mutagenesis. Phosphorylation at S1444 is strikingly reduced in the major PD-related LRRK2 mutations R1441C/G/H, which are part of a consensus PKA recognition site ((1441)RASpS(1444)). Furthermore, our work establishes S1444 as a PKA-regulated 14-3-3 docking site. Experiments of direct binding to the three 14-3-3 isotypes gamma, theta, and zeta with phosphopeptides encompassing pS910, pS935, or pS1444 demonstrated the highest affinities to phospho-S1444. Strikingly, 14-3-3 binding to phospho-S1444 decreased LRRK2 kinase activity in vitro. Moreover, substitution of S1444 by alanine or by introducing the mutations R1441C/G/H, abrogating PKA phosphorylation and 14-3-3 binding, resulted in increased LRRK2 kinase activity. In conclusion, these data clearly demonstrate that LRRK2 kinase activity is modulated by PKA-mediated binding of 14-3-3 to S1444 and suggest that 14-3-3 interaction with LRRK2 is hampered in R1441C/G/H-mediated PD pathogenesis.
Collapse
|
36
|
Taymans JM, Gao F, Baekelandt V. Metabolic labeling of leucine rich repeat kinases 1 and 2 with radioactive phosphate. J Vis Exp 2013:e50523. [PMID: 24084685 DOI: 10.3791/50523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Leucine rich repeat kinases 1 and 2 (LRRK1 and LRRK2) are paralogs which share a similar domain organization, including a serine-threonine kinase domain, a Ras of complex proteins domain (ROC), a C-terminal of ROC domain (COR), and leucine-rich and ankyrin-like repeats at the N-terminus. The precise cellular roles of LRRK1 and LRRK2 have yet to be elucidated, however LRRK1 has been implicated in tyrosine kinase receptor signaling, while LRRK2 is implicated in the pathogenesis of Parkinson's disease. In this report, we present a protocol to label the LRRK1 and LRRK2 proteins in cells with (32)P orthophosphate, thereby providing a means to measure the overall phosphorylation levels of these 2 proteins in cells. In brief, affinity tagged LRRK proteins are expressed in HEK293T cells which are exposed to medium containing (32)P-orthophosphate. The (32)P-orthophosphate is assimilated by the cells after only a few hours of incubation and all molecules in the cell containing phosphates are thereby radioactively labeled. Via the affinity tag (3xflag) the LRRK proteins are isolated from other cellular components by immunoprecipitation. Immunoprecipitates are then separated via SDS-PAGE, blotted to PVDF membranes and analysis of the incorporated phosphates is performed by autoradiography ((32)P signal) and western detection (protein signal) of the proteins on the blots. The protocol can readily be adapted to monitor phosphorylation of any other protein that can be expressed in cells and isolated by immunoprecipitation.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven and Leuven Institute for Neuroscience and Disease (LIND)
| | | | | |
Collapse
|
37
|
Discovery of novel indolinone-based, potent, selective and brain penetrant inhibitors of LRRK2. Bioorg Med Chem Lett 2013; 23:4085-90. [DOI: 10.1016/j.bmcl.2013.05.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/13/2013] [Accepted: 05/15/2013] [Indexed: 12/11/2022]
|
38
|
Ohta E, Kawakami F, Kubo M, Obata F. Dominant-negative effects of LRRK2 heterodimers: a possible mechanism of neurodegeneration in Parkinson's disease caused by LRRK2 I2020T mutation. Biochem Biophys Res Commun 2012; 430:560-6. [PMID: 23220480 DOI: 10.1016/j.bbrc.2012.11.113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 11/28/2012] [Indexed: 10/27/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is the molecule responsible for autosomal-dominant Parkinson's disease (PD), PARK8, but the etiologic effects of its mutation remain unknown. In the present study, we investigated a novel mechanism for the neurodegeneration induced by I2020T mutant LRRK2. Using native gel electrophoresis and immunoprecipitation, we found that wild-type (WT) LRRK2 formed a heterodimer with I2020T LRRK2 in transfected cells, and that the heterodimer exhibited a markedly lower intracellular protein level than the WT/WT-homodimer. An increased amount of I2020T LRRK2 decreased the protein level of co-transfected WT LRRK2. A pulse-chase experiment revealed that the intracellular protein lifetime of WT LRRK2 was shortened by co-transfection with I2020T LRRK2. These results suggest that I2020T LRRK2 enhances the intracellular degradation of WT LRRK2 through WT/I2020T-heterodimer formation. Overexpression of WT LRRK2 in HEK293 cells increased the phosphorylation level of Akt1 (S473), a possible physiological substrate of LRRK2, and made cells resistant to hydrogen peroxide-induced apoptosis. However, both Akt1 phosphorylation and apoptosis resistance were reduced in WT/I2020T-expressing cells in comparison with WT/WT-expressing cells. Reduction of Akt1 phosphorylation and apoptosis resistance were also evident when a neuroblastoma SH-SY5Y clone overexpressing WT LRRK2 was transfected with the I2020T LRRK2. Altogether, these results suggest that the I2020T mutation enhances the intracellular degradation of LRRK2 through WT/I2020T-heterodimer formation, leading to reduced Akt1 phosphorylation and diminished protectivity against apoptosis. Our findings suggest the possibility of a dominant-negative mechanism of neurodegeneration in PD caused by I2020T LRRK2 mutation.
Collapse
Affiliation(s)
- Etsuro Ohta
- Division of Clinical Immunology, Graduate School of Medical Sciences, Kitasato University, Kanagawa, Japan
| | | | | | | |
Collapse
|