1
|
Onat E, Kocaman N, Hançer S, Yildirim M. Role of Dardarin and Isthmin-1 in the Protective Effect of Hydroxytyrosol Against Corn Syrup-Induced Liver Damage. Cureus 2025; 17:e76803. [PMID: 39897276 PMCID: PMC11787043 DOI: 10.7759/cureus.76803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
INTRODUCTION In this study, it was investigated whether dardarin (LRRK2) and isthmin-1 (ISM1) play a role in the protective effect of hydroxytyrosol (HT) used to prevent liver damage caused by corn syrup in rats. METHODS Rats were divided into four groups with six in each group: 1) control, 2) HT, 3) corn syrup, and 4) corn syrup + HT. Rats were given water containing 30% corn syrup for six weeks. At the same time, HT-containing liquid was given orally at 4 ml/kg/day, alone and together with corn syrup for six weeks. The weights of the rats were measured every week. LRRK2 and ISM1 molecules in liver tissue were evaluated by histopathological methods. Biochemical parameters were also examined with the enzyme-linked immunosorbent assay (ELISA) method. RESULTS It was found that weight gain was less in rats receiving HT than in those consuming corn syrup. The increase in cholesterol, triglyceride, and liver enzyme levels because of corn syrup consumption decreased with HT consumption. As a result of histopathological analysis, it was observed that the increase in LRRK2 and ISM1 levels observed in the liver tissue in the corn syrup-administered group decreased when HT was administered together with corn syrup. In addition, it was determined that the increase in sinusoidal expansion and hepatocyte necrosis observed in the liver tissue as a result of corn syrup application decreased as a result of the application of HT together with corn syrup. CONCLUSION The protective effect of HT against the damage caused by corn syrup in the liver has been demonstrated once again; however, LRRK2 and ISM1 are thought to contribute to this issue.
Collapse
Affiliation(s)
- Elif Onat
- Department of Medical Pharmacology, Fırat University, Elazığ, TUR
| | - Nevin Kocaman
- Department of Histology and Embryology, Fırat University, Elazığ, TUR
| | - Serhat Hançer
- Department of Histology and Embryology, Fırat University, Elazığ, TUR
| | - Murat Yildirim
- Department of Pediatrics, University of Wisconsin-Madison, Madison, USA
| |
Collapse
|
2
|
Morez M, Lara Ordóñez AJ, Melnyk P, Liberelle M, Lebègue N, Taymans JM. Leucine-rich repeat kinase 2 (LRRK2) inhibitors for Parkinson's disease: a patent review of the literature to date. Expert Opin Ther Pat 2024; 34:773-788. [PMID: 39023243 DOI: 10.1080/13543776.2024.2378076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/04/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Nearly two decades after leucine rich repeat kinase 2 (LRRK2) was discovered as a genetic determinant of Parkinson's disease (PD), LRRK2 has emerged a priority therapeutic target in PD and inhibition of its activity is hypothesized to be beneficial. AREAS COVERED LRRK2 targeting agents, in particular kinase inhibitors and agents reducing LRRK2 expression show promise in model systems and have progressed to phase I and phase II clinical testing for PD. Several additional targeting strategies for LRRK2 are emerging, based on promoting specific 'healthy' LRRK2 quaternary structures, heteromeric complexes and conformations. EXPERT OPINION It can be expected that LRRK2 targeting strategies may proceed to phase III clinical testing for PD in the next five years, allowing the field to discover the real clinical value of LRRK2 targeting strategies.
Collapse
Affiliation(s)
- Margaux Morez
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | | | - Patricia Melnyk
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Maxime Liberelle
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Nicolas Lebègue
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Jean-Marc Taymans
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| |
Collapse
|
3
|
Tang X, Xing S, Ma M, Xu Z, Guan Q, Chen Y, Feng F, Liu W, Chen T, Chen Y, Sun H. The Development and Design Strategy of Leucine-Rich Repeat Kinase 2 Inhibitors: Promising Therapeutic Agents for Parkinson's Disease. J Med Chem 2023; 66:2282-2307. [PMID: 36758171 DOI: 10.1021/acs.jmedchem.2c01552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting millions of people worldwide. Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are the most common genetic risk factor for PD. Elevated LRRK2 kinase activity is found in idiopathic and familial PD cases. LRRK2 mutations are involved in multiple PD pathogeneses, including dysregulation of mitochondrial homeostasis, ciliogenesis, etc. Here, we provide a comprehensive overview of the biological function, structure, and mutations of LRRK2. We also examine recent advances and challenges in developing LRRK2 inhibitors and address prospective protein-based targeting strategies. The binding mechanisms, structure-activity relationships, and pharmacokinetic features of inhibitors are emphasized to provide a comprehensive compendium on the rational design of LRRK2 inhibitors. We hope that this publication can serve as a guide for designing novel LRRK2 inhibitors based on the summarized facts and perspectives.
Collapse
Affiliation(s)
- Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Mingkang Ma
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Ziwei Xu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qianwen Guan
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuting Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huai'an 223005, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
4
|
Zhao Y, Vavouraki N, Lovering RC, Escott-Price V, Harvey K, Lewis PA, Manzoni C. Tissue specific LRRK2 interactomes reveal a distinct striatal functional unit. PLoS Comput Biol 2023; 19:e1010847. [PMID: 36716346 PMCID: PMC9910798 DOI: 10.1371/journal.pcbi.1010847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 02/09/2023] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Mutations in LRRK2 are the most common genetic cause of Parkinson's disease. Despite substantial research efforts, the physiological and pathological role of this multidomain protein remains poorly defined. In this study, we used a systematic approach to construct the general protein-protein interactome around LRRK2, which was then evaluated taking into consideration the differential expression patterns and the co-expression behaviours of the LRRK2 interactors in 15 different healthy tissue types. The LRRK2 interactors exhibited distinct expression features in the brain as compared to the peripheral tissues analysed. Moreover, a high degree of similarity was found for the LRRK2 interactors in putamen, caudate and nucleus accumbens, thus defining a potential LRRK2 functional cluster within the striatum. The general LRRK2 interactome paired with the expression profiles of its members constitutes a powerful tool to generate tissue-specific LRRK2 interactomes. We exemplified the generation of the tissue-specific LRRK2 interactomes and explored the functions highlighted by the "core LRRK2 interactors" in the striatum in comparison with the cerebellum. Finally, we illustrated how the LRRK2 general interactome reported in this manuscript paired with the expression profiles can be used to trace the relationship between LRRK2 and specific interactors of interest, here focusing on the LRRK2 interactors belonging to the Rab protein family.
Collapse
Affiliation(s)
- Yibo Zhao
- University College London, School of Pharmacy, London, United Kingdom
| | | | - Ruth C. Lovering
- University College London, Institute for Cardiovascular Science, London, United Kingdom
| | - Valentina Escott-Price
- University of Cardiff, School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff, United Kingdom
| | - Kirsten Harvey
- University College London, School of Pharmacy, London, United Kingdom
| | - Patrick A. Lewis
- University of Reading, School of Pharmacy, Reading, United Kingdom
- Royal Veterinary College, London, United Kingdom
- UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Claudia Manzoni
- University College London, School of Pharmacy, London, United Kingdom
| |
Collapse
|
5
|
Filippone A, Mannino D, Cucinotta L, Paterniti I, Esposito E, Campolo M. LRRK2 Inhibition by PF06447475 Antagonist Modulates Early Neuronal Damage after Spinal Cord Trauma. Antioxidants (Basel) 2022; 11:antiox11091634. [PMID: 36139708 PMCID: PMC9495377 DOI: 10.3390/antiox11091634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating event followed by neurodegeneration, activation of the inflammatory cascade, and immune system. The leucine-rich-repeat kinase 2 (LRRK2) is a gene associated with Parkinson’s disease (PD), moreover, its kinase activity was found to be upregulated after instigated inflammation of the central nervous system (CNS). Here, we aimed to investigate the PF06447475 (abbreviated as PF-475) role as a pharmacological LRRK2 antagonist by counteracting pathological consequences of spinal cord trauma. The in vivo model of SCI was induced by extradural compression of the spinal cord, then mice were treated with PF0-475 (2.5–5 and 10 mg/kg i.p) 1 and 6 h after SCI. We found that PF-475 treatments at the higher doses (5 and 10 mg/kg) showed a great ability to significantly reduce the degree of spinal cord tissue injury, glycogen accumulation, and demyelination of neurons associated with trauma. Furthermore, oxidative stress and cytokines expression levels, including interleukins (IL-1, IL-6, IL-10, and 12), interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α), secreted and released after trauma were decreased by LRRK2 antagonist treatments. Our results suggest that the correlations between LRRK2 and inflammation of the CNS exist and that LRRK2 activity targeting could have direct effects on the intervention of neuroinflammatory disorders.
Collapse
|
6
|
Patel A, Patel S, Mehta M, Patel Y, Langaliya D, Bhalodiya S, Bambharoliya T. Recent Update on the Development of Leucine- Rich Repeat Kinase 2 (LRRK2) Inhibitors: A Promising Target for the Treatment of Parkinson's Disease. Med Chem 2022; 18:757-771. [PMID: 35168510 DOI: 10.2174/1573406418666220215122136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease is a relatively common neurological disorder with incidence increasing with age. Since current medications only relieve the symptoms and do not change the course of the disease, therefore, finding disease-modifying therapies is a critical unmet medical need. However, significant progress in understanding how genetics underpins Parkinson's disease (PD) has opened up new opportunities for understanding disease pathogenesis and identifying possible therapeutic targets. One such target is leucine-rich repeat kinase 2 (LRRK2), an elusive enzyme implicated in both familial and idiopathic PD risk. As a result, both academia and industry have promoted the development of potent and selective inhibitors of LRRK2. In this review, we have summarized recent progress on the discovery and development of LRKK2 inhibitors as well as the bioactivity of several small-molecule LRRK2 inhibitors that have been used to inhibit LRRK2 kinase activity in vitro or in vivo.
Collapse
Affiliation(s)
- Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Stuti Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Meshwa Mehta
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Yug Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Dhruv Langaliya
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Shyam Bhalodiya
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | | |
Collapse
|
7
|
Verma A, Ebanks K, Fok CY, Lewis PA, Bettencourt C, Bandopadhyay R. In silico comparative analysis of LRRK2 interactomes from brain, kidney and lung. Brain Res 2021; 1765:147503. [PMID: 33915162 PMCID: PMC8212912 DOI: 10.1016/j.brainres.2021.147503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/06/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023]
Abstract
Mutations in LRRK2 are the most frequent cause of familial Parkinson's disease (PD), with common LRRK2 non-coding variants also acting as risk factors for idiopathic PD. Currently, therapeutic agents targeting LRRK2 are undergoing advanced clinical trials in humans, however, it is important to understand the wider implications of LRRK2 targeted treatments given that LRRK2 is expressed in diverse tissues including the brain, kidney and lungs. This presents challenges to treatment in terms of effects on peripheral organ functioning, thus, protein interactors of LRRK2 could be targeted in lieu to optimize therapeutic effects. Herein an in-silico analysis of LRRK2 direct interactors in brain tissue from various brain regionswas conducted along with a comparative analysis of the LRRK2 interactome in the brain, kidney, and lung tissues. This was carried out based on curated protein-protein interaction (PPI) data from protein interaction databases such as HIPPIE, human gene/protein expression databases and Gene ontology (GO) enrichment analysis using Bingo. Seven targets (MAP2K6, MATK, MAPT, PAK6, SH3GL2, CDC42EP3 and CHGB) were found to be viable objectives for LRRK2 based investigations for PD that would have minimal impact on optimal functioning within peripheral organs. Specifically, MAPT, CHGB, PAK6, and SH3GL2 interacted with LRRK2 in the brain and kidney but not in lung tissue whilst LRRK2-MAP2K6 interacted only in the cerebellum and MATK-LRRK2 interaction was absent in kidney tissues. CDC42EP3 expression levels were low in brain tissues compared to kidney/lung. The results of this computational analysis suggest new avenues for experimental investigations towards LRRK2-targeted therapeutics.
Collapse
Affiliation(s)
- Amrita Verma
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Kirsten Ebanks
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Chi-Yee Fok
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Patrick A Lewis
- Royal Veterinary College, Royal College Street, London NW10TV, United Kingdom; Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Conceicao Bettencourt
- Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom.
| |
Collapse
|
8
|
Azeggagh S, Berwick DC. The development of inhibitors of leucine-rich repeat kinase 2 (LRRK2) as a therapeutic strategy for Parkinson's disease: the current state of play. Br J Pharmacol 2021; 179:1478-1495. [PMID: 34050929 DOI: 10.1111/bph.15575] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/14/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
Current therapeutic approaches for Parkinson's disease (PD) are based around treatments that alleviate symptoms but do not slow or prevent disease progression. As such, alternative strategies are needed. A promising approach is the use of molecules that reduce the function of leucine-rich repeat kinase (LRRK2). Gain-of-function mutations in LRRK2 account for a notable proportion of familial Parkinson's disease cases, and significantly, elevated LRRK2 kinase activity is reported in idiopathic Parkinson's disease. Here, we describe progress in finding therapeutically effective LRRK2 inhibitors, summarising studies that range from in vitro experiments to clinical trials. LRRK2 is a complex protein with two enzymatic activities and a myriad of functions. This creates opportunities for a rich variety of strategies and also increases the risk of unintended consequences. We comment on the strength and limitations of the different approaches and conclude that with two molecules under clinical trial and a diversity of alternative options in the pipeline, there is cause for optimism.
Collapse
Affiliation(s)
- Sonia Azeggagh
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Daniel C Berwick
- Institute of Medical and Biomedical Education, St George's, University of London, London, UK
| |
Collapse
|
9
|
Gloeckner CJ, Porras P. Guilt-by-Association - Functional Insights Gained From Studying the LRRK2 Interactome. Front Neurosci 2020; 14:485. [PMID: 32508578 PMCID: PMC7251075 DOI: 10.3389/fnins.2020.00485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
The Parkinson's disease-associated Leucine-rich repeat kinase 2 (LRRK2) is a complex multi-domain protein belonging to the Roco protein family, a unique group of G-proteins. Variants of this gene are associated with an increased risk of Parkinson's disease. Besides its well-characterized enzymatic activities, conferred by its GTPase and kinase domains, and a central dimerization domain, it contains four predicted repeat domains, which are, based on their structure, commonly involved in protein-protein interactions (PPIs). In the past decades, tremendous progress has been made in determining comprehensive interactome maps for the human proteome. Knowledge of PPIs has been instrumental in assigning functions to proteins involved in human disease and helped to understand the connectivity between different disease pathways and also significantly contributed to the functional understanding of LRRK2. In addition to an increased kinase activity observed for proteins containing PD-associated variants, various studies helped to establish LRRK2 as a large scaffold protein in the interface between cytoskeletal dynamics and the vesicular transport. This review first discusses a number of specific LRRK2-associated PPIs for which a functional consequence can at least be speculated upon, and then considers the representation of LRRK2 protein interactions in public repositories, providing an outlook on open research questions and challenges in this field.
Collapse
Affiliation(s)
- Christian Johannes Gloeckner
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Center for Ophthalmology, Institute for Ophthalmic Research, Core Facility for Medical Bioanalytics, University of Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Pablo Porras
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cherry Hinton, United Kingdom
| |
Collapse
|
10
|
Berwick DC, Heaton GR, Azeggagh S, Harvey K. LRRK2 Biology from structure to dysfunction: research progresses, but the themes remain the same. Mol Neurodegener 2019; 14:49. [PMID: 31864390 PMCID: PMC6925518 DOI: 10.1186/s13024-019-0344-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of leucine-rich repeat kinase 2 (LRRK2) as a protein that is likely central to the aetiology of Parkinson’s disease, a considerable amount of work has gone into uncovering its basic cellular function. This effort has led to the implication of LRRK2 in a bewildering range of cell biological processes and pathways, and probable roles in a number of seemingly unrelated medical conditions. In this review we summarise current knowledge of the basic biochemistry and cellular function of LRRK2. Topics covered include the identification of phosphorylation substrates of LRRK2 kinase activity, in particular Rab proteins, and advances in understanding the activation of LRRK2 kinase activity via dimerisation and association with membranes, especially via interaction with Rab29. We also discuss biochemical studies that shed light on the complex LRRK2 GTPase activity, evidence of roles for LRRK2 in a range of cell signalling pathways that are likely cell type specific, and studies linking LRRK2 to the cell biology of organelles. The latter includes the involvement of LRRK2 in autophagy, endocytosis, and processes at the trans-Golgi network, the endoplasmic reticulum and also key microtubule-based cellular structures. We further propose a mechanism linking LRRK2 dimerisation, GTPase function and membrane recruitment with LRRK2 kinase activation by Rab29. Together these data paint a picture of a research field that in many ways is moving forward with great momentum, but in other ways has not changed fundamentally. Many key advances have been made, but very often they seem to lead back to the same places.
Collapse
Affiliation(s)
- Daniel C Berwick
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK.
| | - George R Heaton
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Sonia Azeggagh
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
11
|
Monteiro LD, Mello FRM, Miranda TP, Heukelbach J. Hansen's disease in children under 15 years old in the state of Tocantins, Brazil, 2001-2012: epidemiological patterns and temporal trends. REVISTA BRASILEIRA DE EPIDEMIOLOGIA 2019; 22:e190047. [PMID: 31460663 DOI: 10.1590/1980-549720190047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/12/2018] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Tocantins is the most hyperendemic state for leprosy in Brazil. OBJECTIVE To describe the epidemiological characteristics and temporal trends of leprosy indicators in children under 15 years old in Tocantins between the years of 2001 and 2012. METHODOLOGY Data analysis of the Notification of Injury Information System (SINAN). New cases under the age of 15 have been included in the state. The indicators were calculated and the temporal trends were analyzed through the join-point regression. RESULTS There were 1,225 cases in children, mean age of 10.8 years, and male predominated (52%). The mode of detection by spontaneous demand prevailed (55.8%) and more than 9% had some physical disability. Detection in < 15 years was significantly increased between 2001 and 2008 (anual percent change - APC = 3.8%; confidence interval of 95% - 95%CI 0.1 - 7.6), and showed significant decline between 2008 and 2012 (APC = -9.4%; 95%CI -17.2 - -0.8). There was stability for the detection of grade 2 cases (APC = 4.2%; 95%CI -6.7 - 16.3), proportion of grade 2 cases (APC = 4.1%; 95%CI 6.7 - 16.3), proportion of grade 1 cases (APC = 1.3%; 95%CI -6.2 - 9.3), multibacillary ratio (APC = 2.9%; 95%CI -1.7 - 7.7), and proportion of paucibacillary (APC = 2.9%; 95%CI -1.7 - 7.7). CONCLUSION Leprosy remains an important public health problem in Tocantins, with active transmission and persistence of transmission foci. The stability of the indicators points out the permanence of the late diagnosis and the repressed demands.
Collapse
Affiliation(s)
- Lorena Dias Monteiro
- Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará - Fortaleza (CE), Brasil.,Fundação Escola de Saúde Pública de Palmas - Palmas (TO), Brasil.,Instituto Tocantinense Presidente Antônio Carlos, ITPAC, Departamento da faculdade de medicina - Palmas (TO), Brasil
| | - Francisco Rogerlândio Martins Mello
- Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará - Fortaleza (CE), Brasil.,Instituto Federal de Educação, Ciência e Tecnologia do Ceará - Caucaia (CE), Brasil
| | | | - Jorg Heukelbach
- Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará - Fortaleza (CE), Brasil.,College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University - Townsville, Queensland, Australia
| |
Collapse
|
12
|
Kozina E, Sadasivan S, Jiao Y, Dou Y, Ma Z, Tan H, Kodali K, Shaw T, Peng J, Smeyne RJ. Mutant LRRK2 mediates peripheral and central immune responses leading to neurodegeneration in vivo. Brain 2019; 141:1753-1769. [PMID: 29800472 DOI: 10.1093/brain/awy077] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Missense mutations in the leucine rich repeat kinase 2 (LRRK2) gene result in late-onset Parkinson's disease. The incomplete penetrance of LRRK2 mutations in humans and LRRK2 murine models of Parkinson's disease suggests that the disease may result from a complex interplay of genetic predispositions and persistent exogenous insults. Since neuroinflammation is commonly associated with the pathogenesis of Parkinson's disease, we examine a potential role of mutant LRRK2 in regulation of the immune response and inflammatory signalling in vivo. Here, we show that mice overexpressing human pathogenic LRRK2 mutations, but not wild-type mice or mice overexpressing human wild-type LRRK2 exhibit long-term lipopolysaccharide-induced nigral neuronal loss. This neurodegeneration is accompanied by an exacerbated neuroinflammation in the brain. The increased immune response in the brain of mutant mice subsequently has an effect on neurons by inducing intraneuronal LRRK2 upregulation. However, the enhanced neuroinflammation is unlikely to be triggered by dysfunctional microglia or infiltrated T cells and/or monocytes, but by peripheral circulating inflammatory molecules. Analysis of cytokine kinetics and inflammatory pathways in the peripheral immune cells demonstrates that LRRK2 mutation alters type II interferon immune response, suggesting that this increased neuroinflammatory response may arise outside the central nervous system. Overall, this study suggests that peripheral immune signalling plays an unexpected-but important-role in the regulation of neurodegeneration in LRRK2-associated Parkinson's disease, and provides new targets for interfering with the onset and progression of the disease.
Collapse
Affiliation(s)
- Elena Kozina
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,Department of Neurosciences, Jefferson Hospital for Neuroscience, Thomas Jefferson University, 900 Walnut St, Philadelphia PA 19107, USA
| | - Shankar Sadasivan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Yun Jiao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Yuchen Dou
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Zhijun Ma
- Department of Hematology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Haiyan Tan
- St. Jude Proteomics Facility, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Kiran Kodali
- St. Jude Proteomics Facility, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Timothy Shaw
- St. Jude Proteomics Facility, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Junmin Peng
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,St. Jude Proteomics Facility, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA
| | - Richard J Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Blvd, Memphis TN 38105, USA.,Department of Neurosciences, Jefferson Hospital for Neuroscience, Thomas Jefferson University, 900 Walnut St, Philadelphia PA 19107, USA
| |
Collapse
|
13
|
Lewis PA. Leucine rich repeat kinase 2: a paradigm for pleiotropy. J Physiol 2019; 597:3511-3521. [PMID: 31124140 DOI: 10.1113/jp276163] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022] Open
Abstract
The LRRK2 gene, coding for leucine rich repeat kinase 2 (LRRK2), is a key player in the genetics of Parkinson's disease. Despite extensive efforts, LRRK2 has proved remarkably evasive with regard to attempts to understand both the role it plays in disease and its normal physiological function. At least part of why LRRK2 has been so difficult to define is that it appears to be many things to many cellular functions and diseases - a pleiotropic actor at both the genetic and the molecular level. Gaining greater insight into the mechanisms and pathways allowing LRRK2 to act in this manner will have implications for our understanding of the role of genes in the aetiology of complex disease, the molecular underpinnings of signal transduction pathways in the cell, and drug discovery in the genome era.
Collapse
Affiliation(s)
- Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
14
|
Rudyk C, Dwyer Z, Hayley S. Leucine-rich repeat kinase-2 (LRRK2) modulates paraquat-induced inflammatory sickness and stress phenotype. J Neuroinflammation 2019; 16:120. [PMID: 31174552 PMCID: PMC6554960 DOI: 10.1186/s12974-019-1483-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/18/2019] [Indexed: 01/11/2023] Open
Abstract
Background Leucine-rich repeat kinase 2 (LRRK2) is a common gene implicated in Parkinson’s disease (PD) and is also thought to be fundamentally involved in numerous immune functions. Thus, we assessed the role of LRRK2 in the context of the effects of the environmental toxicant, paraquat, that has been implicated in PD and is known to affect inflammatory processes. Methods Male LRRK2 knockout (KO) and transgenic mice bearing the G2019S LRRK2 mutation (aged 6–8 months) or their littermate controls were exposed to paraquat (two times per week for 3 weeks), and sickness measures, motivational scores, and total home-cage activity levels were assessed. Following sacrifice, western blot and ELISA assays were performed to see whether or not LRRK2 expression would alter processes related to plasticity, immune response processes, or the stress response. Results Paraquat-induced signs of sickness, inflammation (elevated IL-6), and peripheral toxicity (e.g., organ weight) were completely prevented by LRRK2 knockout. In fact, LRRK2 knockout dramatically reduced not only signs of illness, but also the motivational (nest building) and home-cage activity deficits induced by paraquat. Although LRRK2 deficiency did not affect the striatal BDNF reduction that was provoked by paraquat, it did blunt the corticosterone elevation induced by paraquat, raising the possibility that LRRK2 may modulate aspects of the HPA stress axis. Accordingly, we found that transgenic mice bearing the G2019S LRRK2 mutation had elevated basal corticosterone, along with diminished hippocampal 5-HT1A levels. Conclusion We are the first to show the importance of LRRK2 in the peripheral neurotoxic and stressor-like effects of paraquat. These data are consistent with LRRK2 playing a role in the general inflammatory tone and stressor effects induced by environmental toxicant exposure.
Collapse
Affiliation(s)
- Chris Rudyk
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Zach Dwyer
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada.
| | | |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Mamais A, Manzoni C, Nazish I, Arber C, Sonustun B, Wray S, Warner TT, Cookson MR, Lewis PA, Bandopadhyay R. Analysis of macroautophagy related proteins in G2019S LRRK2 Parkinson's disease brains with Lewy body pathology. Brain Res 2018; 1701:75-84. [PMID: 30055128 PMCID: PMC6361106 DOI: 10.1016/j.brainres.2018.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/04/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
Abstract
LRRK2, the gene encoding the multidomain kinase Leucine-Rich Repeat Kinase 2 (LRRK2), has been linked to familial and sporadic forms of Parkinson's disease (PD), as well as cancer, leprosy and Crohn's disease, establishing it as a target for discovery therapeutics. LRRK2 has been associated with a range of cellular processes, however its physiological and pathological functions remain unclear. The most prevalent LRRK2 mutations in PD have been shown to affect macroautophagy in various cellular models while a role in autophagy signalling has been recapitulated in vivo. Dysregulation of autophagy has been implicated in PD pathology, and this raises the possibility that differential autophagic activity is relevant to disease progression in PD patients carrying LRRK2 mutations. To examine the relevance of LRRK2 to the regulation of macroautophagy in a disease setting we examined the levels of autophagic markers in the basal ganglia of G2019S LRRK2 PD post-mortem tissue, in comparison to pathology-matched idiopathic PD (iPD), using immunoblotting (IB). Significantly lower levels of p62 and LAMP1 were observed in G2019S LRRK2 PD compared to iPD cases. Similarly, an increase in ULK1 was observed in iPD but was not reflected in G2019S LRRK2 PD cases. Furthermore, examination of p62 by immunohistochemistry (IH) recapitulated a distinct signature for G2019S PD. IH of LAMP1, LC3 and ULK1 broadly correlated with the IB results. Our data from a small but pathologically well-characterized cases highlights a divergence of G2019S PD carriers in terms of autophagic response in alpha-synuclein pathology affected brain regions compared to iPD.
Collapse
Affiliation(s)
- Adamantios Mamais
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA.
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom; Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Iqra Nazish
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom
| | - Charles Arber
- Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Berkiye Sonustun
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom
| | - Selina Wray
- Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom; Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom.
| |
Collapse
|
17
|
Tomkins JE, Dihanich S, Beilina A, Ferrari R, Ilacqua N, Cookson MR, Lewis PA, Manzoni C. Comparative Protein Interaction Network Analysis Identifies Shared and Distinct Functions for the Human ROCO Proteins. Proteomics 2018; 18:e1700444. [PMID: 29513927 PMCID: PMC5992104 DOI: 10.1002/pmic.201700444] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/05/2018] [Indexed: 12/19/2022]
Abstract
Signal transduction cascades governed by kinases and GTPases are a critical component of the command and control of cellular processes, with the precise outcome partly determined by direct protein-protein interactions (PPIs). Here, we use the human ROCO proteins as a model for investigating PPI signaling events-taking advantage of the unique dual kinase/GTPase activities and scaffolding properties of these multidomain proteins. PPI networks are reported that encompass the human ROCO proteins, developed using two complementary approaches. First, using the recently developed weighted PPI network analysis (WPPINA) pipeline, a confidence-weighted overview of validated ROCO protein interactors is obtained from peer-reviewed literature. Second, novel ROCO PPIs are assessed experimentally via protein microarray screens. The networks derived from these orthologous approaches are compared to identify common elements within the ROCO protein interactome; functional enrichment analysis of this common core of the network identified stress response and cell projection organization as shared functions within this protein family. Despite the presence of these commonalities, the results suggest that many unique interactors and therefore some specialized cellular roles have evolved for different members of the ROCO proteins. Overall, this multi-approach strategy to increase the resolution of protein interaction networks represents a prototype for the utility of PPI data integration in understanding signaling biology.
Collapse
Affiliation(s)
- James E. Tomkins
- School of PharmacyUniversity of ReadingWhiteknights CampusReadingUK
| | - Sybille Dihanich
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
| | - Alexandra Beilina
- Laboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaUSA
| | - Raffaele Ferrari
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
| | - Nicolò Ilacqua
- School of PharmacyUniversity of ReadingWhiteknights CampusReadingUK
- Department of BiologyUniversity of PadovaPadovaItaly
| | - Mark R. Cookson
- Laboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaUSA
| | - Patrick A. Lewis
- School of PharmacyUniversity of ReadingWhiteknights CampusReadingUK
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
| | - Claudia Manzoni
- School of PharmacyUniversity of ReadingWhiteknights CampusReadingUK
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
| |
Collapse
|
18
|
The LRRK2-macroautophagy axis and its relevance to Parkinson's disease. Biochem Soc Trans 2017; 45:155-162. [PMID: 28202669 PMCID: PMC5310720 DOI: 10.1042/bst20160265] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/04/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
Abstract
A wide variety of different functions and an impressive array of interactors have been associated with leucine-rich repeat kinase 2 (LRRK2) over the years. Here, I discuss the hypothesis that LRRK2 may be capable of interacting with different proteins at different times and places, therefore, controlling a plethora of diverse functions based on the different complexes formed. Among these, I will then focus on macroautophagy in the general context of the endolysosomal system. First, the relevance of autophagy in Parkinson's disease will be evaluated giving a brief overview of all the relevant Parkinson's disease genes; then, the association of LRRK2 with macroautophagy and the endolysosomal pathway will be analyzed based on the supporting literature.
Collapse
|
19
|
Cogo S, Greggio E, Lewis PA. Leucine Rich Repeat Kinase 2: beyond Parkinson's and beyond kinase inhibitors. Expert Opin Ther Targets 2017; 21:751-753. [PMID: 28609155 DOI: 10.1080/14728222.2017.1342968] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Susanna Cogo
- a Department of Biology , University of Padova , Padua , Italy.,b School of Pharmacy , University of Reading , Reading , United Kingdom
| | - Elisa Greggio
- a Department of Biology , University of Padova , Padua , Italy
| | - Patrick A Lewis
- b School of Pharmacy , University of Reading , Reading , United Kingdom.,c Department of Molecular Neuroscience , UCL Institute of Neurology , London , United Kingdom
| |
Collapse
|
20
|
Pathogenic LRRK2 variants are gain-of-function mutations that enhance LRRK2-mediated repression of β-catenin signaling. Mol Neurodegener 2017; 12:9. [PMID: 28103901 PMCID: PMC5248453 DOI: 10.1186/s13024-017-0153-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND LRRK2 mutations and risk variants increase susceptibility to inherited and idiopathic Parkinson's disease, while recent studies have identified potential protective variants. This, and the fact that LRRK2 mutation carriers develop symptoms and brain pathology almost indistinguishable from idiopathic Parkinson's disease, has led to enormous interest in this protein. LRRK2 has been implicated in a range of cellular events, but key among them is canonical Wnt signalling, which results in increased levels of transcriptionally active β-catenin. This pathway is critical for the development and survival of the midbrain dopaminergic neurones typically lost in Parkinson's disease. METHODS Here we use Lrrk2 knockout mice and fibroblasts to investigate the effect of loss of Lrrk2 on canonical Wnt signalling in vitro and in vivo. Micro-computed tomography was used to study predicted tibial strength, while pulldown assays were employed to measure brain β-catenin levels. A combination of luciferase assays, immunofluorescence and co-immunoprecipitation were performed to measure canonical Wnt activity and investigate the relationship between LRRK2 and β-catenin. TOPflash assays are also used to study the effects of LRRK2 kinase inhibition and pathogenic and protective LRRK2 mutations on Wnt signalling. Data were tested by Analysis of Variance. RESULTS Loss of Lrrk2 causes a dose-dependent increase in the levels of transcriptionally active β-catenin in the brain, and alters tibial bone architecture, decreasing the predicted risk of fracture. Lrrk2 knockout cells display increased TOPflash and Axin2 promoter activities, both basally and following Wnt activation. Consistently, over-expressed LRRK2 was found to bind β-catenin and repress TOPflash activation. Some pathogenic LRRK2 mutations and risk variants further suppressed TOPflash, whereas the protective R1398H variant increased Wnt signalling activity. LRRK2 kinase inhibitors affected canonical Wnt signalling differently due to off-targeting; however, specific LRRK2 inhibition reduced canonical Wnt signalling similarly to pathogenic mutations. CONCLUSIONS Loss of LRRK2 causes increased canonical Wnt activity in vitro and in vivo. In agreement, over-expressed LRRK2 binds and represses β-catenin, suggesting LRRK2 may act as part of the β-catenin destruction complex. Since some pathogenic LRRK2 mutations enhance this effect while the protective R1398H variant relieves it, our data strengthen the notion that decreased canonical Wnt activity is central to Parkinson's disease pathogenesis.
Collapse
|
21
|
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been implicated in a wide range of cellular processes, including the catabolic pathways collectively described as autophagy. In this chapter, the evidence linking LRRK2 to autophagy will be examined, along with how regulation of autophagy and lysosomal pathways may provide a nexus between the physiological function of this protein and the different diseases with which it has been associated. Data from cellular and animal models for LRRK2 function and dysfunction support a role in the regulation and control of autophagic pathways in the cell, although the extant results do not provide a clear indication as to whether LRRK2 is a positive or negative regulator of these pathways, and there are conflicting data as to the impact of mutations in LRRK2 causative for Parkinson's disease. Given that LRRK2 is a priority drug target for Parkinson's, the evidence suggesting that knockout or inhibition of LRRK2 can result in deregulation of autophagy may have important implications and is discussed in the context of our wider understanding of LRRK2.
Collapse
Affiliation(s)
- Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK.
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|
22
|
Talari M, Nayak TKS, Kain V, Babu PP, Misra P, Parsa KVL. MicroRNA-712 restrains macrophage pro-inflammatory responses by targeting LRRK2 leading to restoration of insulin stimulated glucose uptake by myoblasts. Mol Immunol 2016; 82:1-9. [PMID: 27992764 DOI: 10.1016/j.molimm.2016.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 02/08/2023]
Abstract
Chronic inflammatory diseases such as insulin resistance, Type 2 diabetes, neurodegenerative diseases etc., are shown to be caused due to imbalanced activation states of macrophages. MicroRNAs which are transcriptional/post-transcriptional regulators of gene expression drive several pathophysiological processes including macrophage polarization. However the functional role of microRNAs in regulating inflammation induced insulin resistance is ill defined. In our current study we observed that the expression of miR-712 was reduced in macrophages exposed to LPS and IFN-γ. Ectopic expression of miR-712 in RAW 264.7 mouse macrophages impaired the expression of iNOS protein and secretion of pro-inflammatory cytokines such as TNF-α, IL-6 and IFN-β which in turn led to improved insulin stimulated glucose uptake in co-cultured L6 myoblasts. Mechanistically, we identified that miR-712 targets the 3'UTR of a potent inflammatory gene LRRK2 and dampens the phosphorylation of p38 and ERK1/2 kinases. Taken together, our data underscore the regulatory role of miR-712 in restoring insulin stimulated glucose uptake by myoblasts through down-regulating macrophage mediated inflammatory responses.
Collapse
Affiliation(s)
- Malathi Talari
- Department of Biology, Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| | - Tapan Kumar Singh Nayak
- Department of Biology, Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| | - Vasundhara Kain
- Department of Biology, Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| | - Phanithi Prakash Babu
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Parimal Misra
- Department of Biology, Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| | - Kishore V L Parsa
- Department of Biology, Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India.
| |
Collapse
|
23
|
L'RRK de Triomphe: a solution for LRRK2 GTPase activity? Biochem Soc Trans 2016; 44:1625-1634. [DOI: 10.1042/bst20160240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/15/2016] [Accepted: 09/23/2016] [Indexed: 01/01/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a central protein in the pathogenesis of Parkinson's disease (PD), yet its normal function has proved stubbornly hard to elucidate. Even though it remains unclear how pathogenic mutations affect LRRK2 to cause PD, recent findings provide increasing cause for optimism. We summarise here the developing consensus over the effect of pathogenic mutations in the Ras of complex proteins and C-terminal of Roc domains on LRRK2 GTPase activity. This body of work has been greatly reinforced by our own study of the protective R1398H variant contained within the LRRK2 GTPase domain. Collectively, data point towards the pathogenicity of GTP-bound LRRK2 and strengthen a working model for LRRK2 GTPase function as a GTPase activated by dimerisation. Together with the identification of the protective R1398H variant as a valuable control for pathogenic mutations, we have no doubt that these triumphs for the LRRK2 field will accelerate research towards resolving LRRK2 function and towards new treatments for PD.
Collapse
|
24
|
Manzoni C, Mamais A, Roosen DA, Dihanich S, Soutar MPM, Plun-Favreau H, Bandopadhyay R, Hardy J, Tooze SA, Cookson MR, Lewis PA. mTOR independent regulation of macroautophagy by Leucine Rich Repeat Kinase 2 via Beclin-1. Sci Rep 2016; 6:35106. [PMID: 27731364 PMCID: PMC5059726 DOI: 10.1038/srep35106] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 09/22/2016] [Indexed: 12/29/2022] Open
Abstract
Leucine rich repeat kinase 2 is a complex enzyme with both kinase and GTPase activities, closely linked to the pathogenesis of several human disorders including Parkinson's disease, Crohn's disease, leprosy and cancer. LRRK2 has been implicated in numerous cellular processes; however its physiological function remains unclear. Recent reports suggest that LRRK2 can act to regulate the cellular catabolic process of macroautophagy, although the precise mechanism whereby this occurs has not been identified. To investigate the signalling events through which LRRK2 acts to influence macroautophagy, the mammalian target of rapamycin (mTOR)/Unc-51-like kinase 1 (ULK1) and Beclin-1/phosphatidylinositol 3-kinase (PI3K) pathways were evaluated in astrocytic cell models in the presence and absence of LRRK2 kinase inhibitors. Chemical inhibition of LRRK2 kinase activity resulted in the stimulation of macroautophagy in a non-canonical fashion, independent of mTOR and ULK1, but dependent upon the activation of Beclin 1-containing class III PI3-kinase.
Collapse
Affiliation(s)
- Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, United Kingdom
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Dorien A. Roosen
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Sybille Dihanich
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Marc P. M. Soutar
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Helene Plun-Favreau
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street London WC1N 1PJ, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Sharon A. Tooze
- Francis Crick Institute, London Research Institute, 44 Lincoln’s Inn Fields London, WC2A 3LY, United Kingdom
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Patrick A. Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, United Kingdom
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| |
Collapse
|
25
|
Kuwahara T, Inoue K, D’Agati VD, Fujimoto T, Eguchi T, Saha S, Wolozin B, Iwatsubo T, Abeliovich A. LRRK2 and RAB7L1 coordinately regulate axonal morphology and lysosome integrity in diverse cellular contexts. Sci Rep 2016; 6:29945. [PMID: 27424887 PMCID: PMC4947924 DOI: 10.1038/srep29945] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been linked to several clinical disorders including Parkinson's disease (PD), Crohn's disease, and leprosy. Furthermore in rodents, LRRK2 deficiency or inhibition leads to lysosomal pathology in kidney and lung. Here we provide evidence that LRRK2 functions together with a second PD-associated gene, RAB7L1, within an evolutionarily conserved genetic module in diverse cellular contexts. In C. elegans neurons, orthologues of LRRK2 and RAB7L1 act coordinately in an ordered genetic pathway to regulate axonal elongation. Further genetic studies implicated the AP-3 complex, which is a known regulator of axonal morphology as well as of intracellular protein trafficking to the lysosome compartment, as a physiological downstream effector of LRRK2 and RAB7L1. Additional cell-based studies implicated LRRK2 in the AP-3 complex-related intracellular trafficking of lysosomal membrane proteins. In mice, deficiency of either RAB7L1 or LRRK2 leads to prominent age-associated lysosomal defects in kidney proximal tubule cells, in the absence of frank CNS pathology. We hypothesize that defects in this evolutionarily conserved genetic pathway underlie the diverse pathologies associated with LRRK2 in humans and in animal models.
Collapse
Affiliation(s)
- Tomoki Kuwahara
- Departments of Pathology, Cell Biology and Neurology, and Taub Institute, Columbia University, New York, NY, 10032, USA
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Keiichi Inoue
- Departments of Pathology, Cell Biology and Neurology, and Taub Institute, Columbia University, New York, NY, 10032, USA
| | - Vivette D. D’Agati
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Tetta Fujimoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Tomoya Eguchi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shamol Saha
- Department of Pharmacology and Experimental Therapeutics and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Asa Abeliovich
- Departments of Pathology, Cell Biology and Neurology, and Taub Institute, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
26
|
Fava VM, Manry J, Cobat A, Orlova M, Van Thuc N, Ba NN, Thai VH, Abel L, Alcaïs A, Schurr E. A Missense LRRK2 Variant Is a Risk Factor for Excessive Inflammatory Responses in Leprosy. PLoS Negl Trop Dis 2016; 10:e0004412. [PMID: 26844546 PMCID: PMC4742274 DOI: 10.1371/journal.pntd.0004412] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/08/2016] [Indexed: 12/17/2022] Open
Abstract
Background Depending on the epidemiological setting, a variable proportion of leprosy patients will suffer from excessive pro-inflammatory responses, termed type-1 reactions (T1R). The LRRK2 gene encodes a multi-functional protein that has been shown to modulate pro-inflammatory responses. Variants near the LRRK2 gene have been associated with leprosy in some but not in other studies. We hypothesized that LRRK2 was a T1R susceptibility gene and that inconsistent association results might reflect different proportions of patients with T1R in the different sample settings. Hence, we evaluated the association of LRRK2 variants with T1R susceptibility. Methodology An association scan of the LRRK2 locus was performed using 156 single-nucleotide polymorphisms (SNPs). Evidence of association was evaluated in two family-based samples: A set of T1R-affected and a second set of T1R-free families. Only SNPs significant for T1R-affected families with significant evidence of heterogeneity relative to T1R-free families were considered T1R-specific. An expression quantitative trait locus (eQTL) analysis was applied to evaluate the impact of T1R-specific SNPs on LRRK2 gene transcriptional levels. Principal Findings A total of 18 T1R-specific variants organized in four bins were detected. The core SNP capturing the T1R association was the LRRK2 missense variant M2397T (rs3761863) that affects LRRK2 protein turnover. Additionally, a bin of nine SNPs associated with T1R were eQTLs for LRRK2 in unstimulated whole blood cells but not after exposure to Mycobacterium leprae antigen. Significance The results support a preferential association of LRRK2 variants with T1R. LRRK2 involvement in T1R is likely due to a pathological pro-inflammatory loop modulated by LRRK2 availability. Interestingly, the M2397T variant was reported in association with Crohn’s disease with the same risk allele as in T1R suggesting common inflammatory mechanism in these two distinct diseases. A major challenge of current leprosy control is the management of host pathological immune responses coined Type-1 Reactions (T1R). T1R are characterized by acute inflammatory episodes whereby cellular immune responses are directed against host peripheral nerve cells. T1R affects up half of all leprosy patients and are a major cause of leprosy-associated disabilities. Since there is evidence that host genetic factors predispose leprosy patients to T1R, we have conducted a candidate gene study to test if LRRK2 gene variants are T1R risk factors. The choice of LRRK2 was motivated by the fact that LRRK2 was associated with leprosy per se in some but not in other studies. We reasoned that this may reflect different proportions of leprosy patients with T1R in the different samples and that LRRK2 may in truth be a T1R susceptibility gene. Here, we show that variants overlapping the LRRK2 gene, reported as suggestive leprosy per se susceptibility factors in a previous genome-wide association study, are preferentially associated with T1R. The main SNP carrying most of the association signal is the amino-acid change M2397T (rs3761863) which is known to impact LRRK2 turnover. Interestingly, eQTL SNPs counterbalanced the effect of the M2397T variant but this compensatory mechanism was abrogated by Mycobacterium leprae antigen stimulation.
Collapse
Affiliation(s)
- Vinicius M. Fava
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Canada
| | - Jérémy Manry
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Canada
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
| | - Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Canada
| | | | - Nguyen Ngoc Ba
- Hospital for Dermato-Venerology, Ho Chi Minh City, Vietnam
| | - Vu Hong Thai
- Hospital for Dermato-Venerology, Ho Chi Minh City, Vietnam
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, United States of America
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, United States of America
- Centre d’Investigation Clinique, Unité de Recherche Clinique, Necker and Cochin Hospitals, Paris, France
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Canada
- * E-mail:
| | | |
Collapse
|
27
|
Abstract
The last 2 decades represent a period of unparalleled advancement in the understanding of the pathogenesis of Parkinson disease (PD). The discovery of several forms of familial parkinsonism with mendelian inheritance has elucidated insights into the mechanisms underlying the degeneration of dopaminergic neurons of the substantia nigra that histologically characterize PD. α-Synuclein, the principal component of Lewy bodies, remains the presumed pathogen at the heart of the current model; however, concurrently, a diverse range of other mechanisms have been implicated. The creation of a coherent disease model will be crucial to the development of effective disease modifying therapies for sporadic PD.
Collapse
Affiliation(s)
- Stephen Mullin
- Department of Clinical Neurosciences, UCL Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical Neurosciences, UCL Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK.
| |
Collapse
|
28
|
Abstract
BACKGROUND Parkinson's disease (PD) was previously described as the prototypical sporadic disease; however, rapid advances in population and molecular genetics have revealed the existence of a significant number genetic risk factors, prompting its redefinition as a primarily genetic disorder. SOURCES OF DATA Data for this review have been gathered from the published literature. AREAS OF AGREEMENT Multiple haplotypes conveying variable but quantifiable genetic risk, acting concurrently and possibly interacting with one another, provide the basis for a new model of PD. The beginning of this revolution in our understanding came from the clinical observation of parkinsonism with a Mendelian pattern of inheritance in a number of families. The functional work that followed elucidated multiple disease pathways leading to the degeneration of the substantia nigra that characterizes PD. It is however only in recent years, with the emergence of large cohort genome-wide association studies (GWAS), that the relevance of these pathways to so-called sporadic PD has become apparent. AREAS OF CONTROVERSY A substantial portion of the presumed genetic inheritance of PD remains at present undefined. Although it is likely that so-called intermediate risk genetic risk factors are the principal component of this 'missing heritability', this is yet to be proved. GROWING POINTS Although the picture is by now means complete, the beginnings of rational basis for genetic screening of PD risk have begun to emerge. Equally, this enhanced understanding of the various genetic and in turn biochemical pathways shows promising signs of producing fruitful therapeutic strategies. Technological advances promise to reduce the costs associated with and further increase our capability to understand the complex influence of genetics on the pathogenesis of PD. AREAS TIMELY FOR DEVELOPING RESEARCH The coming years will require the enhancement of current techniques and the development of new ones to define PD's missing heritability. It will also require functional work to define better and in turn potentially reverse the mechanisms that contribute with large effect sizes to the risk of sporadic PD.
Collapse
Affiliation(s)
- Stephen Mullin
- Leonard Wolfson Clinical Research Fellow, UCL, Institute of Neurology, Rowland Hill Street, Hampstead, London NW3 2PF, UK
| | - Anthony Schapira
- Department of Clinical Neurosciences, UCL, Institute of Neurology, Hampstead, London, UK
| |
Collapse
|
29
|
Chao YX, Chew LM, Deng X, Tan EK. Nonmotor symptoms in sporadic versus familial forms of Parkinson's disease. Neurodegener Dis Manag 2015; 5:147-53. [DOI: 10.2217/nmt.14.57] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
SUMMARY Besides the classical motor symptoms, Parkinson's disease (PD) patients experience a wide range of nonmotor symptoms (NMS) throughout the disease course. However, due to the lack of recognition and understanding of the pathogenesis, NMS symptoms may be overlooked. Familial PD is a well-defined group that can provide a good model to investigate the mechanisms for both motor and NMS in PD. Some studies suggest that the frequency of NMS is not different between genetic and sporadic form of PD while others suggest that specific domains (such as neuropsychiatric symptoms) are more common in the genetic form. Early recognition of NMS may facilitate early diagnosis and monitoring of both sporadic and genetic PD.
Collapse
Affiliation(s)
- Yin Xia Chao
- National Neuroscience Institute, Singapore 308433, Singapore
- Duke-National University of Singapore Graduate Medical School, Singapore 169857, Singapore
| | - Lai Mun Chew
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Neurology, Singapore General Hospital, Singapore 169608, Singapore
| | - Xiao Deng
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Neurology, Singapore General Hospital, Singapore 169608, Singapore
| | - Eng-King Tan
- National Neuroscience Institute, Singapore 308433, Singapore
- Duke-National University of Singapore Graduate Medical School, Singapore 169857, Singapore
- Department of Neurology, Singapore General Hospital, Singapore 169608, Singapore
| |
Collapse
|
30
|
Progressive dopaminergic alterations and mitochondrial abnormalities in LRRK2 G2019S knock-in mice. Neurobiol Dis 2015; 78:172-95. [PMID: 25836420 DOI: 10.1016/j.nbd.2015.02.031] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/19/2015] [Accepted: 02/21/2015] [Indexed: 01/19/2023] Open
Abstract
Mutations in the LRRK2 gene represent the most common genetic cause of late onset Parkinson's disease. The physiological and pathological roles of LRRK2 are yet to be fully determined but evidence points towards LRRK2 mutations causing a gain in kinase function, impacting on neuronal maintenance, vesicular dynamics and neurotransmitter release. To explore the role of physiological levels of mutant LRRK2, we created knock-in (KI) mice harboring the most common LRRK2 mutation G2019S in their own genome. We have performed comprehensive dopaminergic, behavioral and neuropathological analyses in this model up to 24months of age. We find elevated kinase activity in the brain of both heterozygous and homozygous mice. Although normal at 6months, by 12months of age, basal and pharmacologically induced extracellular release of dopamine is impaired in both heterozygous and homozygous mice, corroborating previous findings in transgenic models over-expressing mutant LRRK2. Via in vivo microdialysis measurement of basal and drug-evoked extracellular release of dopamine and its metabolites, our findings indicate that exocytotic release from the vesicular pool is impaired. Furthermore, profound mitochondrial abnormalities are evident in the striatum of older homozygous G2019S KI mice, which are consistent with mitochondrial fission arrest. We anticipate that this G2019S mouse line will be a useful pre-clinical model for further evaluation of early mechanistic events in LRRK2 pathogenesis and for second-hit approaches to model disease progression.
Collapse
|
31
|
Manzoni C, Denny P, Lovering RC, Lewis PA. Computational analysis of the LRRK2 interactome. PeerJ 2015; 3:e778. [PMID: 25737818 PMCID: PMC4338795 DOI: 10.7717/peerj.778] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/29/2015] [Indexed: 12/13/2022] Open
Abstract
LRRK2 was identified in 2004 as the causative protein product of the Parkinson’s disease locus designated PARK8. In the decade since then, genetic studies have revealed at least 6 dominant mutations in LRRK2 linked to Parkinson’s disease, alongside one associated with cancer. It is now well established that coding changes in LRRK2 are one of the most common causes of Parkinson’s. Genome-wide association studies (GWAs) have, more recently, reported single nucleotide polymorphisms (SNPs) around the LRRK2 locus to be associated with risk of developing sporadic Parkinson’s disease and inflammatory bowel disorder. The functional research that has followed these genetic breakthroughs has generated an extensive literature regarding LRRK2 pathophysiology; however, there is still no consensus as to the biological function of LRRK2. To provide insight into the aspects of cell biology that are consistently related to LRRK2 activity, we analysed the plethora of candidate LRRK2 interactors available through the BioGRID and IntAct data repositories. We then performed GO terms enrichment for the LRRK2 interactome. We found that, in two different enrichment portals, the LRRK2 interactome was associated with terms referring to transport, cellular organization, vesicles and the cytoskeleton. We also verified that 21 of the LRRK2 interactors are genetically linked to risk for Parkinson’s disease or inflammatory bowel disorder. The implications of these findings are discussed, with particular regard to potential novel areas of investigation.
Collapse
Affiliation(s)
- Claudia Manzoni
- School of Pharmacy, University of Reading , Whiteknights, Reading , UK ; Department of Molecular Neuroscience, UCL Institute of Neurology, University College London , Queen Square, London , UK
| | - Paul Denny
- Institute of Cardiovascular Science, University College London , London , UK
| | - Ruth C Lovering
- Institute of Cardiovascular Science, University College London , London , UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading , Whiteknights, Reading , UK ; Department of Molecular Neuroscience, UCL Institute of Neurology, University College London , Queen Square, London , UK ; Centre for Integrated Neuroscience and Neurodynamics, University of Reading , Whiteknights, Reading , UK
| |
Collapse
|
32
|
Insights into the molecular mechanisms of action of bioportides: a strategy to target protein-protein interactions. Expert Rev Mol Med 2015; 17:e1. [DOI: 10.1017/erm.2014.24] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell-penetrating peptides (CPPs) are reliable vehicles for the target-selective intracellular delivery of therapeutic agents. The identification and application of numerous intrinsically bioactive CPPs, now designated as bioportides, is further endorsement of the tremendous clinical potential of CPP technologies. The refinement of proteomimetic bioportides, particularly sequences that mimic cationic α-helical domains involved in protein-protein interactions (PPIs), provides tremendous opportunities to modulate this emergent drug modality in a clinical setting. Thus, a number of CPP-based constructs are currently undergoing clinical trials as human therapeutics, with a particular focus upon anti-cancer agents. A well-characterised array of synthetic modifications, compatible with modern solid-phase synthesis, can be utilised to improve the biophysical and pharmacological properties of bioportides and so achieve cell-and tissue-selective targeting in vivo. Moreover, considering the recent successful development of stapled α-helical peptides as anti-cancer agents, we hypothesise that similar structural modifications are applicable to the design of bioportides that more effectively modulate the many interactomes known to underlie human diseases. Thus, we propose that stapled-helical bioportides could satisfy all of the clinical requirements for metabolically stable, intrinsically cell-permeable agents capable of regulating discrete PPIs by a dominant negative mode of action with minimal toxicity.
Collapse
|
33
|
Wang D, Xu L, Lv L, Su LY, Fan Y, Zhang DF, Bi R, Yu D, Zhang W, Li XA, Li YY, Yao YG. Association of the LRRK2 genetic polymorphisms with leprosy in Han Chinese from Southwest China. Genes Immun 2014; 16:112-9. [PMID: 25521227 DOI: 10.1038/gene.2014.72] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/19/2014] [Accepted: 10/27/2014] [Indexed: 12/31/2022]
Abstract
Leprosy is a chronic infectious and neurological disease that is caused by infection of Mycobacterium leprae (M. leprae). A recent genome-wide association study indicated a suggestive association of LRRK2 genetic variant rs1873613 with leprosy in Chinese population. To validate this association and further identify potential causal variants of LRRK2 with leprosy, we genotyped 13 LRRK2 variants in 548 leprosy patients and 1078 healthy individuals from Yunnan Province and (re-)analyzed 3225 Han Chinese across China. Variants rs1427267, rs3761863, rs1873613, rs732374 and rs7298930 were significantly associated with leprosy per se and/or paucibacillary leprosy (PB). Haplotype A-G-A-C-A was significantly associated with leprosy per se (P=0.018) and PB (P=0.020). Overexpression of the protective allele (Thr2397) of rs3761863 in HEK293 cells led to a significantly increased nuclear factor of activated T-cells' activity compared with allele Met2397 after lipopolysaccharides stimulation. Allele Thr2397 could attenuate 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced autophagic activity in U251 cells. These data suggest that the protective effect of LRRK2 variant p.M2397T on leprosy might be mediated by increasing immune response and decreasing neurotoxicity after M. leprae loading. Our findings confirm that LRRK2 is a susceptible gene to leprosy in Han Chinese population.
Collapse
Affiliation(s)
- D Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - L Xu
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - L Lv
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - L-Y Su
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Y Fan
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - D-F Zhang
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - R Bi
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China [2] Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - D Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - W Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - X-A Li
- Yuxi City Center for Disease Control and Prevention, Yuxi, Yunnan, China
| | - Y-Y Li
- Department of Dermatology, the First Affiliated Hospital of Kunming Medical College, Kunming, Yunnan, China
| | - Y-G Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
34
|
Recent advances in Parkinson’s disease genetics. J Neurol 2014; 261:259-66. [PMID: 23798000 DOI: 10.1007/s00415-013-7003-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/07/2013] [Accepted: 06/08/2013] [Indexed: 12/15/2022]
Abstract
The last 5 years have seen rapid progress in Parkinson’s disease (PD) genetics, with the publication of a series of large-scale genome wide association studies for PD, and evaluation of the roles of the LRRK2 and GBA genes in the aetiology of PD. We are beginning to develop a coherent picture of the interplay of Mendelian and non-Mendelian factors in PD. Pathways involved in mitochondrial quality control (mitophagy), lysosomal function and immune function are emerging as important in the pathogenesis of PD. These pathways represent a target for therapeutic intervention and a way in which the heterogeneity of disease cause, as well as disease mechanism, can be established. In the future, there is likely to be an individualised basis for the treatment of PD, linked to the identification of specific genetic factors.
Collapse
|
35
|
Genetic, structural, and molecular insights into the function of ras of complex proteins domains. ACTA ACUST UNITED AC 2014; 21:809-18. [PMID: 24981771 PMCID: PMC4104024 DOI: 10.1016/j.chembiol.2014.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/13/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
Abstract
Ras of complex proteins (ROC) domains were identified in 2003 as GTP binding modules in large multidomain proteins from Dictyostelium discoideum. Research into the function of these domains exploded with their identification in a number of proteins linked to human disease, including leucine-rich repeat kinase 2 (LRRK2) and death-associated protein kinase 1 (DAPK1) in Parkinson’s disease and cancer, respectively. This surge in research has resulted in a growing body of data revealing the role that ROC domains play in regulating protein function and signaling pathways. In this review, recent advances in the structural information available for proteins containing ROC domains, along with insights into enzymatic function and the integration of ROC domains as molecular switches in a cellular and organismal context, are explored.
Collapse
|
36
|
Kethiri RR, Bakthavatchalam R. Leucine-rich repeat kinase 2 inhibitors: a review of recent patents (2011 – 2013). Expert Opin Ther Pat 2014; 24:745-57. [DOI: 10.1517/13543776.2014.907275] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
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: 40] [Impact Index Per Article: 3.6] [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
|
38
|
Jiang E, Li F, Jing C, Li P, Cui H, Wang B, Ding M, Pang H. High-Resolution Melting Analysis as a Developed Method for Genotyping the PD Susceptibility Loci in LRRK2 Gene. J Clin Lab Anal 2014; 29:299-304. [PMID: 24849765 DOI: 10.1002/jcla.21769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 04/02/2014] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Single-nucleotide polymorphisms (SNPs) have been reported as a highly relevant point for the mechanisms of Parkinson's disease (PD). The invention of saturating dye makes it possible to identify heteroduplex DNA without redistribution during melting, which allows using high-resolution melting (HRM) to detect SNPs. However, the HRM analysis for detection of those SNPs associated with PD was rarely applied. METHODS Two SNPs, G2385R and R1628P, located in leucine-rich repeat kinase 2 (LRRK2) gene were individually and multiplexedly genotyped using HRM analysis. The sequence variant observed in unexpected HRM curves was confirmed by DNA sequencing. RESULTS HRM analysis identified successfully all genotypes both on R1628P and G2385R loci. The unexpected HRM curves appeared in R1628P amplicon generated from combinations of R1628P and rs11176013 loci. A multiplexed HRM assay that genotyped R1628P, rs11176013, and G2385R loci was efficiently established. CONCLUSIONS The present HRM assay is a reliable and rapid method for genotyping R1628P and G2385R loci in LRRK2 gene, and multiplex HRM analysis results in high throughput and has the potential to facilitate a wide range of genotyping studies on PD susceptibility genes.
Collapse
Affiliation(s)
- Enzhu Jiang
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Fengrui Li
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
- Department of Forensic Medicine, Baotou Medical College, Baotou, P.R. China
| | - Chenchen Jing
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Pei Li
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Honggang Cui
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Baojie Wang
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Mei Ding
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| | - Hao Pang
- School of Forensic Medicine, China Medical University, Shenyang, P.R. China
| |
Collapse
|
39
|
No association between genetic variants of the LRRK2 gene and schizophrenia in Han Chinese. Neurosci Lett 2014; 566:210-5. [PMID: 24631561 DOI: 10.1016/j.neulet.2014.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/10/2014] [Accepted: 03/04/2014] [Indexed: 12/18/2022]
Abstract
Mitochondrial dysfunction was widely reported in schizophrenia patients in recent studies. Leucine-rich repeat kinase 2 (LRRK2) is a mitochondrial protein, and mutations in the LRRK2 gene can induce mitochondrial dysfunction. LRRK2 mutations have been reported to be the most frequent genetic cause of Parkinson's disease (PD). We were interested in whether LRRK2 variants also play a role in schizophrenia. In this study, we genotyped 12 genetic variants (including 4 tag SNPs and 8 disease-associated variants) in the LRRK2 gene in a total of 2449 samples composed of two independent Han Chinese schizophrenia case-control cohorts (486 schizophrenia patients and 480 healthy controls from Hunan Province; 624 schizophrenia patients and 859 healthy controls from Shanghai). We compared the genotype, allele and haplotype frequencies of those SNPs between cases and controls. Statistical analyses revealed no association between LRRK2 variants/haplotypes and schizophrenia in these two schizophrenia case-control cohorts and the combined samples. Our results indicated that the LRRK2 variants are unlikely to be actively involved in schizophrenia in Han Chinese.
Collapse
|
40
|
Mutant LRRK2 toxicity in neurons depends on LRRK2 levels and synuclein but not kinase activity or inclusion bodies. J Neurosci 2014; 34:418-33. [PMID: 24403142 DOI: 10.1523/jneurosci.2712-13.2014] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
By combining experimental neuron models and mathematical tools, we developed a "systems" approach to deconvolve cellular mechanisms of neurodegeneration underlying the most common known cause of Parkinson's disease (PD), mutations in leucine-rich repeat kinase 2 (LRRK2). Neurons ectopically expressing mutant LRRK2 formed inclusion bodies (IBs), retracted neurites, accumulated synuclein, and died prematurely, recapitulating key features of PD. Degeneration was predicted from the levels of diffuse mutant LRRK2 that each neuron contained, but IB formation was neither necessary nor sufficient for death. Genetic or pharmacological blockade of its kinase activity destabilized LRRK2 and lowered its levels enough to account for the moderate reduction in LRRK2 toxicity that ensued. By contrast, targeting synuclein, including neurons made from PD patient-derived induced pluripotent cells, dramatically reduced LRRK2-dependent neurodegeneration and LRRK2 levels. These findings suggest that LRRK2 levels are more important than kinase activity per se in predicting toxicity and implicate synuclein as a major mediator of LRRK2-induced neurodegeneration.
Collapse
|
41
|
Abstract
Wingless/Int (Wnt) signaling pathways are signal transduction mechanisms that have been widely studied in the field of embryogenesis. Recent work has established a critical role for these pathways in brain development, especially of midbrain dopaminergic neurones. However, the fundamental importance of Wnt signaling for the normal function of mature neurones in the adult central nervous system has also lately been demonstrated by an increasing number of studies. Parkinson's disease (PD) is the second most prevalent neurodegenerative disease worldwide and is currently incurable. This debilitating disease is characterized by the progressive loss of a subset of midbrain dopaminergic neurones in the substantia nigra leading to typical extrapyramidal motor symptoms. The aetiology of PD is poorly understood but work performed over the last two decades has identified a growing number of genetic defects that underlie this condition. Here we review a growing body of data connecting genes implicated in PD--most notably the PARK genes--with Wnt signaling. These observations provide clues to the normal function of these proteins in healthy neurones and suggest that deregulated Wnt signaling might be a frequent pathomechanism leading to PD. These observations have implications for the pathogenesis and treatment of neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Daniel C. Berwick
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| |
Collapse
|
42
|
Estrada AA, Chan BK, Baker-Glenn C, Beresford A, Burdick DJ, Chambers M, Chen H, Dominguez SL, Dotson J, Drummond J, Flagella M, Fuji R, Gill A, Halladay J, Harris SF, Heffron TP, Kleinheinz T, Lee DW, Pichon CEL, Liu X, Lyssikatos JP, Medhurst AD, Moffat JG, Nash K, Scearce-Levie K, Sheng Z, Shore DG, Wong S, Zhang S, Zhang X, Zhu H, Sweeney ZK. Discovery of Highly Potent, Selective, and Brain-Penetrant Aminopyrazole Leucine-Rich Repeat Kinase 2 (LRRK2) Small Molecule Inhibitors. J Med Chem 2014; 57:921-36. [DOI: 10.1021/jm401654j] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Anthony A. Estrada
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Bryan K. Chan
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Charles Baker-Glenn
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Alan Beresford
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Daniel J. Burdick
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Mark Chambers
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Huifen Chen
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Sara L. Dominguez
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Jennafer Dotson
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Jason Drummond
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Michael Flagella
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Reina Fuji
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Andrew Gill
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Jason Halladay
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Seth F. Harris
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Timothy P. Heffron
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Tracy Kleinheinz
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Donna W. Lee
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Claire E. Le Pichon
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Xingrong Liu
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Joseph P. Lyssikatos
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Andrew D. Medhurst
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - John G. Moffat
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Kevin Nash
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Kimberly Scearce-Levie
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Zejuan Sheng
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Daniel G. Shore
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Susan Wong
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Shuo Zhang
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Xiaolin Zhang
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Haitao Zhu
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| | - Zachary K. Sweeney
- Departments of †Discovery Chemistry, ‡Neurosciences, §Biochemical and Cellular
Pharmacology, ∥Drug Metabolism
and Pharmacokinetics, ⊥Safety Assessment, and #Structural Biology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
- Departments
of ∇Chemistry, ○Biochemical and
Cellular Pharmacology, and ◆Drug Metabolism
and Pharmacokinetics, BioFocus, Chesterford Research Park, Saffron Walden, CB10 1XL, United Kingdom
| |
Collapse
|
43
|
Zhu H, Chen H, Cho W, Estrada AA, Sweeney ZK. From Human Genetics to Drug Candidates: An Industrial Perspective on LRRK2 Inhibition as a Treatment for Parkinson's Disease. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2013. [DOI: 10.1002/9783527677252.ch10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
44
|
Manzoni C, Mamais A, Dihanich S, Abeti R, Soutar MPM, Plun-Favreau H, Giunti P, Tooze SA, Bandopadhyay R, Lewis PA. Inhibition of LRRK2 kinase activity stimulates macroautophagy. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:2900-2910. [PMID: 23916833 PMCID: PMC3898616 DOI: 10.1016/j.bbamcr.2013.07.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 02/05/2023]
Abstract
Leucine Rich Repeat Kinase 2 (LRRK2) is one of the most important genetic contributors to Parkinson's disease. LRRK2 has been implicated in a number of cellular processes, including macroautophagy. To test whether LRRK2 has a role in regulating autophagy, a specific inhibitor of the kinase activity of LRRK2 was applied to human neuroglioma cells and downstream readouts of autophagy examined. The resulting data demonstrate that inhibition of LRRK2 kinase activity stimulates macroautophagy in the absence of any alteration in the translational targets of mTORC1, suggesting that LRRK2 regulates autophagic vesicle formation independent of canonical mTORC1 signaling. This study represents the first pharmacological dissection of the role LRRK2 plays in the autophagy/lysosomal pathway, emphasizing the importance of this pathway as a marker for LRRK2 physiological function. Moreover it highlights the need to dissect autophagy and lysosomal activities in the context of LRRK2 related pathologies with the final aim of understanding their aetiology and identifying specific targets for disease modifying therapies in patients.
Collapse
Affiliation(s)
- Claudia Manzoni
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| | - Adamantios Mamais
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK; Reta Lila Weston Institute and Queen Square Brain Bank, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Sybille Dihanich
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Rosella Abeti
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Marc P M Soutar
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Helene Plun-Favreau
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Paola Giunti
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sharon A Tooze
- London Research Institute, Cancer Research UK, Lincoln's Inn Fields, London, WC2A 3LY, UK
| | - Rina Bandopadhyay
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK; Reta Lila Weston Institute and Queen Square Brain Bank, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Patrick A Lewis
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK; School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
45
|
Manzoni C, Mamais A, Dihanich S, McGoldrick P, Devine MJ, Zerle J, Kara E, Taanman JW, Healy DG, Marti-Masso JF, Schapira AH, Plun-Favreau H, Tooze S, Hardy J, Bandopadhyay R, Lewis PA. Pathogenic Parkinson's disease mutations across the functional domains of LRRK2 alter the autophagic/lysosomal response to starvation. Biochem Biophys Res Commun 2013; 441:862-6. [PMID: 24211199 PMCID: PMC3858825 DOI: 10.1016/j.bbrc.2013.10.159] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 10/29/2013] [Indexed: 11/16/2022]
Abstract
LRRK2 is one of the most important genetic contributors to Parkinson's disease (PD). Point mutations in this gene cause an autosomal dominant form of PD, but to date no cellular phenotype has been consistently linked with mutations in each of the functional domains (ROC, COR and Kinase) of the protein product of this gene. In this study, primary fibroblasts from individuals carrying pathogenic mutations in the three central domains of LRRK2 were assessed for alterations in the autophagy/lysosomal pathway using a combination of biochemical and cellular approaches. Mutations in all three domains resulted in alterations in markers for autophagy/lysosomal function compared to wild type cells. These data highlight the autophagy and lysosomal pathways as read outs for pathogenic LRRK2 function and as a marker for disease, and provide insight into the mechanisms linking LRRK2 function and mutations.
Collapse
Affiliation(s)
- Claudia Manzoni
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Adamantios Mamais
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Sybille Dihanich
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Phillip McGoldrick
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Michael J. Devine
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Julia Zerle
- Helmholtz Zentrum München, GmbH Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| | - Eleanna Kara
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jan-Willem Taanman
- Department of Clinical Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Daniel G. Healy
- Beaumont Hospital, 9 Beaumont Rd, Dublin 9, Co. Dublin, Ireland
| | | | - Anthony H. Schapira
- Department of Clinical Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Helene Plun-Favreau
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Sharon Tooze
- London Research Institute, Cancer Research UK, London WC2A 3LY, UK
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Rina Bandopadhyay
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Patrick A. Lewis
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AP, UK
| |
Collapse
|
46
|
Dihanich S, Civiero L, Manzoni C, Mamais A, Bandopadhyay R, Greggio E, Lewis PA. GTP binding controls complex formation by the human ROCO protein MASL1. FEBS J 2013; 281:261-74. [PMID: 24286120 PMCID: PMC3995007 DOI: 10.1111/febs.12593] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 10/22/2013] [Accepted: 10/28/2013] [Indexed: 12/12/2022]
Abstract
The human ROCO proteins are a family of multi-domain proteins sharing a conserved ROC-COR supra-domain. The family has four members: leucine-rich repeat kinase 1 (LRRK1), leucine-rich repeat kinase 2 (LRRK2), death-associated protein kinase 1 (DAPK1) and malignant fibrous histiocytoma amplified sequences with leucine-rich tandem repeats 1 (MASL1). Previous studies of LRRK1/2 and DAPK1 have shown that the ROC (Ras of complex proteins) domain can bind and hydrolyse GTP, but the cellular consequences of this activity are still unclear. Here, the first biochemical characterization of MASL1 and the impact of GTP binding on MASL1 complex formation are reported. The results demonstrate that MASL1, similar to other ROCO proteins, can bind guanosine nucleotides via its ROC domain. Furthermore, MASL1 exists in two distinct cellular complexes associated with heat shock protein 60, and the formation of a low molecular weight pool of MASL1 is modulated by GTP binding. Finally, loss of GTP enhances MASL1 toxicity in cells. Taken together, these data point to a central role for the ROC/GTPase domain of MASL1 in the regulation of its cellular function.
Collapse
Affiliation(s)
- Sybille Dihanich
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | | | | | | | | | | |
Collapse
|
47
|
Marcinek P, Jha AN, Shinde V, Sundaramoorthy A, Rajkumar R, Suryadevara NC, Neela SK, van Tong H, Balachander V, Valluri VL, Thangaraj K, Velavan TP. LRRK2 and RIPK2 variants in the NOD 2-mediated signaling pathway are associated with susceptibility to Mycobacterium leprae in Indian populations. PLoS One 2013; 8:e73103. [PMID: 24015287 PMCID: PMC3756038 DOI: 10.1371/journal.pone.0073103] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/24/2013] [Indexed: 01/14/2023] Open
Abstract
In recent years, genome wide association studies have discovered a large number of gene loci that play a functional role in innate and adaptive immune pathways associated with leprosy susceptibility. The immunological control of intracellular bacteria M. leprae is modulated by NOD2-mediated signaling of Th1 responses. In this study, we investigated 211 clinically classified leprosy patients and 230 ethnically matched controls in Indian population by genotyping four variants in NOD2 (rs9302752A/G), LRRK2 (rs1873613A/G), RIPK2 (rs40457A/G and rs42490G/A). The LRRK2 locus is associated with leprosy outcome. The LRRK2 rs1873613A minor allele and respective rs1873613AA genotypes were significantly associated with an increased risk whereas the LRRK2 rs1873613G major allele and rs1873613GG genotypes confer protection in paucibacillary and leprosy patients. The reconstructed GA haplotypes from RIPK2 rs40457A/G and rs42490G/A variants was observed to contribute towards increased risk whereas haplotypes AA was observed to confer protective role. Our results indicate that a possible shared mechanisms underlying the development of these two clinical forms of the disease as hypothesized. Our findings confirm and validates the role of gene variants involved in NOD2-mediated signalling pathways that play a role in immunological control of intracellular bacteria M. leprae.
Collapse
Affiliation(s)
- Patrick Marcinek
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Trabzuni D, Ryten M, Emmett W, Ramasamy A, Lackner KJ, Zeller T, Walker R, Smith C, Lewis PA, Mamais A, de Silva R, Vandrovcova J, Hernandez D, Nalls MA, Sharma M, Garnier S, Lesage S, Simon-Sanchez J, Gasser T, Heutink P, Brice A, Singleton A, Cai H, Schadt E, Wood NW, Bandopadhyay R, Weale ME, Hardy J, Plagnol V. Fine-mapping, gene expression and splicing analysis of the disease associated LRRK2 locus. PLoS One 2013; 8:e70724. [PMID: 23967090 PMCID: PMC3742662 DOI: 10.1371/journal.pone.0070724] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 06/23/2013] [Indexed: 12/04/2022] Open
Abstract
Association studies have identified several signals at the LRRK2 locus for Parkinson's disease (PD), Crohn's disease (CD) and leprosy. However, little is known about the molecular mechanisms mediating these effects. To further characterize this locus, we fine-mapped the risk association in 5,802 PD and 5,556 controls using a dense genotyping array (ImmunoChip). Using samples from 134 post-mortem control adult human brains (UK Human Brain Expression Consortium), where up to ten brain regions were available per individual, we studied the regional variation, splicing and regulation of LRRK2. We found convincing evidence for a common variant PD association located outside of the LRRK2 protein coding region (rs117762348, A>G, P = 2.56×10(-8), case/control MAF 0.083/0.074, odds ratio 0.86 for the minor allele with 95% confidence interval [0.80-0.91]). We show that mRNA expression levels are highest in cortical regions and lowest in cerebellum. We find an exon quantitative trait locus (QTL) in brain samples that localizes to exons 32-33 and investigate the molecular basis of this eQTL using RNA-Seq data in n = 8 brain samples. The genotype underlying this eQTL is in strong linkage disequilibrium with the CD associated non-synonymous SNP rs3761863 (M2397T). We found two additional QTLs in liver and monocyte samples but none of these explained the common variant PD association at rs117762348. Our results characterize the LRRK2 locus, and highlight the importance and difficulties of fine-mapping and integration of multiple datasets to delineate pathogenic variants and thus develop an understanding of disease mechanisms.
Collapse
Affiliation(s)
- Daniah Trabzuni
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mina Ryten
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Warren Emmett
- University College London Genetics Institute, University College London, London, United Kingdom
| | - Adaikalavan Ramasamy
- Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Mainz, Germany
| | - Tanja Zeller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany
| | - Robert Walker
- MRC Sudden Death Brain Bank Project, University of Edinburgh, Department of Neuropathology, Edinburgh, Scotland, United Kingdom
| | - Colin Smith
- MRC Sudden Death Brain Bank Project, University of Edinburgh, Department of Neuropathology, Edinburgh, Scotland, United Kingdom
| | - Patrick A. Lewis
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- School of Pharmacy, University of Reading, Whiteknights, Reading, United Kingdom
| | - Adamantios Mamais
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Reta Lila Weston Institute of Neurological Studies, London, United Kingdom
| | - Rohan de Silva
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Reta Lila Weston Institute of Neurological Studies, London, United Kingdom
| | - Jana Vandrovcova
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Reta Lila Weston Institute of Neurological Studies, London, United Kingdom
| | | | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Manu Sharma
- Division of Neurodegenerative Disorders, Hertie Institute for Clinical Brain Research, University of Tubingen, Tubingen, Germany
| | - Sophie Garnier
- Pierre and Marie Curie University, Institut National de la Santé et de la Recherche Médicale UMRS 937, Paris, France
| | - Suzanne Lesage
- CRICM, University Pierre et Marie Curie, Institut National de la Santé et de la Recherche Médicale UMRS 975, CNRS UMR 7225, Hospital Pitié-Salpêtrière, Paris, France
| | - Javier Simon-Sanchez
- Department of Clinical Genetics, Section of Medical Genomics, VU University Medical Centre, Amsterdam, The Netherlands
| | - Thomas Gasser
- Division of Neurodegenerative Disorders, Hertie Institute for Clinical Brain Research, University of Tubingen, Tubingen, Germany
| | - Peter Heutink
- Department of Clinical Genetics, Section of Medical Genomics, VU University Medical Centre, Amsterdam, The Netherlands
| | - Alexis Brice
- CRICM, University Pierre et Marie Curie, Institut National de la Santé et de la Recherche Médicale UMRS 975, CNRS UMR 7225, Hospital Pitié-Salpêtrière, Paris, France
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Huaibin Cai
- Unit of Transgenesis, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eric Schadt
- Institute for Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Nicholas W. Wood
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Rina Bandopadhyay
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Reta Lila Weston Institute of Neurological Studies, London, United Kingdom
| | - Michael E. Weale
- Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
- Reta Lila Weston Institute of Neurological Studies, London, United Kingdom
| | - Vincent Plagnol
- University College London Genetics Institute, University College London, London, United Kingdom
| |
Collapse
|
49
|
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: 2.4] [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]
|
50
|
Berwick DC, Harvey K. LRRK2: an éminence grise of Wnt-mediated neurogenesis? Front Cell Neurosci 2013; 7:82. [PMID: 23754980 PMCID: PMC3668263 DOI: 10.3389/fncel.2013.00082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/13/2013] [Indexed: 01/13/2023] Open
Abstract
The importance of leucine-rich repeat kinase 2 (LRRK2) to mature neurons is well-established, since mutations in PARK8, the gene encoding LRRK2, are the most common known cause of Parkinson’s disease. Nonetheless, despite the LRRK2 knockout mouse having no overt neurodevelopmental defect, numerous lines of in vitro data point toward a central role for this protein in neurogenesis. Roles for LRRK2 have been described in many key processes, including neurite outgrowth and the regulation of microtubule dynamics. Moreover, LRRK2 has been implicated in cell cycle control, suggesting additional roles in neurogenesis that precede terminal differentiation. However, we contend that the suggested function of LRRK2 as a scaffolding protein at the heart of numerous Wnt signaling cascades provides the most tantalizing link to neurogenesis in the developing brain. Numerous lines of evidence show a critical requirement for multiple Wnt pathways in the development of certain brain regions, not least the dopaminergic neurons of the ventral mid-brain. In conclusion, these observations indicate a function of LRRK2 as a subtle yet critical mediator of the action of Wnt ligands on developing neurons. We suggest that LRRK2 loss- or gain-of-function are likely modifiers of developmental phenotypes seen in animal models of Wnt signaling deregulation, a hypothesis that can be tested by cross-breeding relevant genetically modified experimental strains.
Collapse
Affiliation(s)
- Daniel C Berwick
- Department of Pharmacology, University College London School of Pharmacy, University College London London, UK
| | | |
Collapse
|