101
|
Neefjes J, Jongsma MML, Berlin I. Stop or Go? Endosome Positioning in the Establishment of Compartment Architecture, Dynamics, and Function. Trends Cell Biol 2017; 27:580-594. [PMID: 28363667 DOI: 10.1016/j.tcb.2017.03.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 02/03/2023]
Abstract
The endosomal system constitutes a key negotiator between the environment of a cell and its internal affairs. Comprised of a complex membranous network, wherein each vesicle can in principle move autonomously throughout the cell, the endosomal system operates as a coherent unit to optimally face external challenges and maintain homeostasis. Our appreciation of how individual endosomes are controlled in time and space to best serve their collective purpose has evolved dramatically in recent years. In light of these efforts, the endoplasmic reticulum (ER) - with its expanse of membranes permeating the cytoplasmic space - has emerged as a potent spatiotemporal organizer of endosome biology. We review the latest advances in our understanding of the mechanisms underpinning endosomal transport and positioning, with emphasis on the contributions from the ER, and offer a perspective on how the interplay between these aspects shapes the architecture and dynamics of the endosomal system and drives its myriad cellular functions.
Collapse
Affiliation(s)
- Jacques Neefjes
- Department of Chemical Immunology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Marlieke M L Jongsma
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC)/Universiteit van Amsterdam (UvA), Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Ilana Berlin
- Department of Chemical Immunology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| |
Collapse
|
102
|
Pathogenic Huntington Alters BMP Signaling and Synaptic Growth through Local Disruptions of Endosomal Compartments. J Neurosci 2017; 37:3425-3439. [PMID: 28235896 DOI: 10.1523/jneurosci.2752-16.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 01/05/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a polyglutamine (polyQ) stretch within the Huntingtin (Htt) protein. Pathogenic Htt disrupts multiple neuronal processes, including gene expression, axonal trafficking, proteasome and mitochondrial activity, and intracellular vesicle trafficking. However, the primary pathogenic mechanism and subcellular site of action for mutant Htt are still unclear. Using a Drosophila HD model, we found that pathogenic Htt expression leads to a profound overgrowth of synaptic connections that correlates directly with the levels of Htt at nerve terminals. Branches of the same nerve containing different levels of Htt show distinct phenotypes, indicating that Htt acts locally to disrupt synaptic growth. The effects of pathogenic Htt on synaptic growth arise from defective synaptic endosomal trafficking, leading to expansion of a recycling endosomal signaling compartment containing Sorting Nexin 16 and a reduction in late endosomes containing Rab11. The disruption of endosomal compartments leads to elevated BMP signaling within nerve terminals, driving excessive synaptic growth. Blocking aberrant signaling from endosomes or reducing BMP activity ameliorates the severity of HD pathology and improves viability. Pathogenic Htt is present largely in a nonaggregated form at synapses, indicating that cytosolic forms of the protein are likely to be the toxic species that disrupt endosomal signaling. Our data indicate that pathogenic Htt acts locally at nerve terminals to alter trafficking between endosomal compartments, leading to defects in synaptic structure that correlate with pathogenesis and lethality in the Drosophila HD model.SIGNIFICANCE STATEMENT Huntington's disease (HD) is the most commonly inherited polyglutamine expansion disorder, but how mutant Huntingtin (Htt) disrupts neuronal function is unclear. In particular, it is unknown within what subcellular compartment pathogenic Htt acts and whether the pathogenesis is associated with aggregated or more soluble forms of the protein. Using a Drosophila HD model, we find that nonaggregated pathogenic Htt acts locally at synaptic terminals to disrupt endosomal compartments, leading to aberrant wiring defects. Genetic manipulations to increase endosomal trafficking of synaptic growth receptors from signaling endosomes or to reduce BMP signaling reduce pathology in this HD model. These data indicate that pathogenic Htt can act locally within nerve terminals to disrupt synaptic endosomal signaling and induce neuropathology.
Collapse
|
103
|
Maas JWJ, Yang J, Edwards RH. Endogenous Leucine-Rich Repeat Kinase 2 Slows Synaptic Vesicle Recycling in Striatal Neurons. Front Synaptic Neurosci 2017; 9:5. [PMID: 28280464 PMCID: PMC5322269 DOI: 10.3389/fnsyn.2017.00005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/10/2017] [Indexed: 12/27/2022] Open
Abstract
Dominant mutations in leucine-rich repeat kinase 2 (LRRK2) produce the most common inherited form of Parkinson’s disease (PD) but the function of LRRK2 remains poorly understood. The presynaptic role of multiple genes linked to PD including α-synuclein (α-syn) has suggested that LRRK2 may also influence neurotransmitter release, a possibility supported by recent work. However, the use of disease-associated mutants that cause toxicity complicates the analysis. To determine whether LRRK2 normally influences the synaptic vesicle, we have now used a combination of imaging and electrophysiology to study LRRK2 knockout (KO) mice. Surprisingly, we find that in hippocampal (generally excitatory) neurons, the loss of LRRK2 does not affect synaptic vesicle exocytosis, endocytosis or the mobility of α-syn. Double KO (DKO) mice lacking LRRK1 as well as LRRK2 also show no defect in transmitter release by hippocampal neurons. However, in striatal neurons, which express LRRK2 at higher levels, the loss of LRRK2 leads to modest acceleration of synaptic vesicle endocytosis. Thus, endogenous LRRK2 normally slows synaptic vesicle recycling at striatal terminals.
Collapse
Affiliation(s)
- James W Jr Maas
- Departments of Neurology and Physiology, Weill Institute for Neurosciences, and Kavli Institute for Fundamental Neuroscience, UCSF School of Medicine San Francisco, CA, USA
| | - Jing Yang
- Departments of Neurology and Physiology, Weill Institute for Neurosciences, and Kavli Institute for Fundamental Neuroscience, UCSF School of Medicine San Francisco, CA, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, Weill Institute for Neurosciences, and Kavli Institute for Fundamental Neuroscience, UCSF School of Medicine San Francisco, CA, USA
| |
Collapse
|
104
|
Zhang Y, Schmid B, Nikolaisen NK, Rasmussen MA, Aldana BI, Agger M, Calloe K, Stummann TC, Larsen HM, Nielsen TT, Huang J, Xu F, Liu X, Bolund L, Meyer M, Bak LK, Waagepetersen HS, Luo Y, Nielsen JE, Holst B, Clausen C, Hyttel P, Freude KK. Patient iPSC-Derived Neurons for Disease Modeling of Frontotemporal Dementia with Mutation in CHMP2B. Stem Cell Reports 2017; 8:648-658. [PMID: 28216144 PMCID: PMC5355623 DOI: 10.1016/j.stemcr.2017.01.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022] Open
Abstract
The truncated mutant form of the charged multivesicular body protein 2B (CHMP2B) is causative for frontotemporal dementia linked to chromosome 3 (FTD3). CHMP2B is a constituent of the endosomal sorting complex required for transport (ESCRT) and, when mutated, disrupts endosome-to-lysosome trafficking and substrate degradation. To understand the underlying molecular pathology, FTD3 patient induced pluripotent stem cells (iPSCs) were differentiated into forebrain-type cortical neurons. FTD3 neurons exhibited abnormal endosomes, as previously shown in patients. Moreover, mitochondria of FTD3 neurons displayed defective cristae formation, accompanied by deficiencies in mitochondrial respiration and increased levels of reactive oxygen. In addition, we provide evidence for perturbed iron homeostasis, presenting an in vitro patient-specific model to study the effects of iron accumulation in neurodegenerative diseases. All phenotypes observed in FTD3 neurons were rescued in CRISPR/Cas9-edited isogenic controls. These findings illustrate the relevance of our patient-specific in vitro models and open up possibilities for drug target development. FTD3 neurons show abnormalities in endosomes and mitochondria Parkinson's and Alzheimer's disease core genes are altered in FTD3 neurons Iron homeostasis is perturbed in FTD3 neurons Impairments in FTD3 neurons are rescued in CRISPR/Cas9-edited isogenic controls
Collapse
Affiliation(s)
- Yu Zhang
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark.
| | | | - Nanett K Nikolaisen
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | | | - Blanca I Aldana
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mikkel Agger
- Stem Cell and Developmental Neurobiology Group, Department of Neurobiology Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Kirstine Calloe
- The Physiology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | | | - Hjalte M Larsen
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Troels T Nielsen
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jinrong Huang
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Fengping Xu
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Xin Liu
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Lars Bolund
- Danish Regenerative Engineering Alliance for Medicine (DREAM), Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Morten Meyer
- Stem Cell and Developmental Neurobiology Group, Department of Neurobiology Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lasse K Bak
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Helle S Waagepetersen
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Yonglun Luo
- Danish Regenerative Engineering Alliance for Medicine (DREAM), Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jørgen E Nielsen
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | | | | | - Poul Hyttel
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Kristine K Freude
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark.
| |
Collapse
|
105
|
Martin I. Decoding Parkinson's Disease Pathogenesis: The Role of Deregulated mRNA Translation. JOURNAL OF PARKINSONS DISEASE 2017; 6:17-27. [PMID: 26889638 PMCID: PMC4927901 DOI: 10.3233/jpd-150738] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mutations in a number of genes cause rare familial forms of Parkinson’s disease and provide profound insight into potential mechanisms governing disease pathogenesis. Recently, a role for translation and metabolism of mRNA has emerged in the development of various neurodegenerative disorders including Parkinson’s disease (PD). In PD, preliminary evidence supports a role for aberrant translation in the disease process stemming from mutations in several genes. Translation control is central to maintaining organism homeostasis under variable environmental conditions and deregulation of this may predispose to certain stressors. Hypothetically, deregulated translation may be detrimental to neuronal viability in PD through the misexpression of a subset of transcripts or through the impact of excessive bulk translation on energy consumption and burden on protein homeostatic mechanisms. While compelling preliminary evidence exists to support a role for translation in PD, much more work is required to identify specific mechanisms linking altered translation to the disease process.
Collapse
Affiliation(s)
- Ian Martin
- Correspondence to: Ian Martin, PhD Jungers Center for Neurosciences Research Parkinson Center of Oregon Department of Neurology - Mail Code L623 Oregon Health and Science University 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA. Tel.: +1 503 494 9140; Fax: +1 503 494 7358; E-mail:
| |
Collapse
|
106
|
Thomas JM, Li T, Yang W, Xue F, Fishman PS, Smith WW. 68 and FX2149 Attenuate Mutant LRRK2-R1441C-Induced Neural Transport Impairment. Front Aging Neurosci 2017; 8:337. [PMID: 28119604 PMCID: PMC5222795 DOI: 10.3389/fnagi.2016.00337] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/26/2016] [Indexed: 11/27/2022] Open
Abstract
Leucine-rich repeat kinase 2 is a large protein with implications in genetic and sporadic causes of Parkinson's disease. The physiological functions of LRRK2 are largely unknown. In this report, we investigated whether LRRK2 alters neural transport using live-cell imaging techniques and human neuroblastoma SH-SY5Y cells. Our results demonstrated that expression of the PD-linked mutant, LRRK2-R1441C, induced mitochondrial, and lysosomal transport defects in neurites of SH-SY5Y cells. Most importantly, recently identified GTP-binding inhibitors, 68 and FX2149, can reduce LRRK2 GTP-binding activity and attenuates R1441C-induced mitochondrial and lysosomal transport impairments. These results provide direct evidence and an early mechanism for neurite injury underlying LRRK2-induced neurodegeneration. This is the first report to show that LRRK2 GTP-binding activity plays a critical role during neurite transport, suggesting inhibition of LRRK2 GTP-binding could be a potential novel strategy for PD intervention.
Collapse
Affiliation(s)
- Joseph M Thomas
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore, MD, USA
| | - Tianxia Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore, MD, USA
| | - Wei Yang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore, MD, USA
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore, MD, USA
| | - Paul S Fishman
- Department of Neurology, University of Maryland School of MedicineBaltimore, MD, USA; Neurology Service, VA Maryland Healthcare SystemBaltimore, MD, USA
| | - Wanli W Smith
- Department of Psychiatry, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
107
|
Xiong Y, Dawson TM, Dawson VL. Models of LRRK2-Associated Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2017; 14:163-191. [PMID: 28353284 DOI: 10.1007/978-3-319-49969-7_9] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic causes of Parkinson's disease (PD) and also one of the strongest genetic risk factors in sporadic PD. The LRRK2 protein contains a GTPase and a kinase domain and several protein-protein interaction domains. Both in vitro and in vivo assays in different model systems have provided tremendous insights into the molecular mechanisms underlying LRRK2-induced dopaminergic neurodegeneration. Among all the model systems, animal models are crucial tools to study the pathogenesis of human disease. How do the animal models recapitulate LRRK2-induced dopaminergic neuronal loss in human PD? To answer this question, this review focuses on the discussion of the animal models of LRRK2-associated PD including genetic- and viral-based models.
Collapse
Affiliation(s)
- Yulan Xiong
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Anatomy and Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS, 66506, USA.
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, 70130-2685, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, 70130-2685, USA. .,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
108
|
Roosen DA, Cookson MR. LRRK2 at the interface of autophagosomes, endosomes and lysosomes. Mol Neurodegener 2016; 11:73. [PMID: 27927216 PMCID: PMC5142374 DOI: 10.1186/s13024-016-0140-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023] Open
Abstract
Over the past 20 years, substantial progress has been made in identifying the underlying genetics of Parkinson's disease (PD). Of the known genes, LRRK2 is a major genetic contributor to PD. However, the exact function of LRRK2 remains to be elucidated. In this review, we discuss how familial forms of PD have led us to hypothesize that alterations in endomembrane trafficking play a role in the pathobiology of PD. We will discuss the major observations that have been made to elucidate the role of LRRK2 in particular, including LRRK2 animal models and high-throughput proteomics approaches. Taken together, these studies strongly support a role of LRRK2 in vesicular dynamics. We also propose that targeting these pathways may not only be beneficial for developing therapeutics for LRRK2-driven PD, but also for other familial and sporadic cases.
Collapse
Affiliation(s)
- Dorien A. Roosen
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP UK
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
| |
Collapse
|
109
|
Lloyd-Evans E, Haslett LJ. The lysosomal storage disease continuum with ageing-related neurodegenerative disease. Ageing Res Rev 2016; 32:104-121. [PMID: 27516378 DOI: 10.1016/j.arr.2016.07.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 12/11/2022]
Abstract
Lysosomal storage diseases and diseases of ageing share many features both at the physiological level and with respect to the mechanisms that underlie disease pathogenesis. Although the exact pathophysiology is not exactly the same, it is astounding how many similar pathways are altered in all of these diseases. The aim of this review is to provide a summary of the shared disease mechanisms, outlining the similarities and differences and how genetics, insight into rare diseases and functional research has changed our perspective on the causes underlying common diseases of ageing. The lysosome should no longer be considered as just the stomach of the cell or as a suicide bag, it has an emerging role in cellular signalling, nutrient sensing and recycling. The lysosome is of fundamental importance in the pathophysiology of diseases of ageing and by comparing against the LSDs we not only identify common pathways but also therapeutic targets so that ultimately more effective treatments can be developed for all neurodegenerative diseases.
Collapse
|
110
|
Pu J, Guardia CM, Keren-Kaplan T, Bonifacino JS. Mechanisms and functions of lysosome positioning. J Cell Sci 2016; 129:4329-4339. [PMID: 27799357 DOI: 10.1242/jcs.196287] [Citation(s) in RCA: 319] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysosomes have been classically considered terminal degradative organelles, but in recent years they have been found to participate in many other cellular processes, including killing of intracellular pathogens, antigen presentation, plasma membrane repair, cell adhesion and migration, tumor invasion and metastasis, apoptotic cell death, metabolic signaling and gene regulation. In addition, lysosome dysfunction has been shown to underlie not only rare lysosome storage disorders but also more common diseases, such as cancer and neurodegeneration. The involvement of lysosomes in most of these processes is now known to depend on the ability of lysosomes to move throughout the cytoplasm. Here, we review recent findings on the mechanisms that mediate the motility and positioning of lysosomes, and the importance of lysosome dynamics for cell physiology and pathology.
Collapse
Affiliation(s)
- Jing Pu
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tal Keren-Kaplan
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
111
|
Atashrazm F, Dzamko N. LRRK2 inhibitors and their potential in the treatment of Parkinson's disease: current perspectives. Clin Pharmacol 2016; 8:177-189. [PMID: 27799832 PMCID: PMC5076802 DOI: 10.2147/cpaa.s102191] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Major advances in understanding how genetics underlies Parkinson's disease (PD) have provided new opportunities for understanding disease pathogenesis and potential new targets for therapeutic intervention. One such target is leucine-rich repeat kinase 2 (LRRK2), an enigmatic enzyme implicated in both familial and idiopathic PD risk. Both academia and industry have promoted the development of potent and selective inhibitors of LRRK2, and these are currently being employed to assess the safety and efficacy of such compounds in preclinical models of PD. This review examines the evidence that LRRK2 kinase activity contributes to the pathogenesis of PD and outlines recent progress on inhibitor development and early results from preclinical safety and efficacy testing. This review also looks at some of the challenges remaining for translation of LRRK2 inhibitors to the clinic, if indeed this is ultimately warranted. As a disease with no current cure that is increasing in prevalence in line with an aging population, there is much need for developing new treatments for PD, and targeting LRRK2 is currently a promising option.
Collapse
Affiliation(s)
| | - Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|
112
|
Abstract
The power of Drosophila genetics has attracted attention in tackling important biomedical challenges such as the understanding and prevention of neurodegenerative diseases. Parkinson's disease (PD) is the most common neurodegenerative movement disorder which results from the relentless degeneration of midbrain dopaminergic neurons. Over the past two decades tremendous advances have been made in identifying genes responsible for inherited forms of PD. The ease of genetic manipulation in Drosophila has spurred the development of numerous models of PD, including expression of human genes carrying pathogenic mutations or the targeted mutation of conserved orthologs. The genetic and cellular analysis of these models is beginning to reveal fundamental insights into the pathogenic mechanisms. Numerous pathways and processes are disrupted in these models but some common themes are emerging. These often implicate aberrant synaptic function, protein aggregation, autophagy, oxidative stress, and mitochondrial dysfunction. Moreover, an impressive list of small molecule compounds have been identified as effective in reversing pathogenic phenotypes, paving the way to explore these for therapeutic interventions.
Collapse
Affiliation(s)
- V L Hewitt
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom
| | - A J Whitworth
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom.
| |
Collapse
|
113
|
Esteves AR, Cardoso SM. LRRK2 at the Crossroad Between Autophagy and Microtubule Trafficking: Insights into Parkinson's Disease. Neuroscientist 2016; 23:16-26. [PMID: 26740081 DOI: 10.1177/1073858415616558] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mutations in leucine-rich repeat kinase 2 ( lrrk2) gene cause inherited Parkinson's disease (PD), and common variants in lrrk2 are a risk factor for sporadic PD. The neuropathology associated with LRRK2-linked PD is extremely pleomorphic involving inclusions of α-synuclein (SNCA), tau or neither, therefore suggesting that LRRK2 may be central in the pathogenic pathways of PD. This large protein localizes in the cytosol, as well as, in specific membrane domains, including mitochondria and autophagosomes and interacts with a wide range of proteins such as SNCA, tau, α- and β-tubulin. For this reason LRRK2 has been associated with a variety of cellular functions, including autophagy, mitochondrial function/dynamics and microtubule/cytoskeletal dynamics. LRRK2 has been shown to interact with microtubules as well as with mitochondria interfering with their network and dynamics. Moreover, LRRK2 knock-out or mutations affect autophagic efficiency. Here, we review and discuss the literature on how LRRK2 affects mitochondrial function, autophagy, and microtubule dynamics and how this is implicated in the PD etiology.
Collapse
Affiliation(s)
- A Raquel Esteves
- 1 CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,2 CNC-IBILI, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- 1 CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,2 CNC-IBILI, University of Coimbra, Coimbra, Portugal.,3 Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
114
|
Ma B, Xu L, Pan X, Sun L, Ding J, Xie C, Koliatsos VE, Cai H. LRRK2 modulates microglial activity through regulation of chemokine (C-X3-C) receptor 1 -mediated signalling pathways. Hum Mol Genet 2016; 25:3515-3523. [PMID: 27378696 DOI: 10.1093/hmg/ddw194] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/14/2016] [Accepted: 06/16/2016] [Indexed: 12/15/2022] Open
Abstract
Multiple missense mutations in Leucine-rich repeat kinase 2 (LRRK2) have been linked to Parkinson's disease (PD), the most common degenerative movement disorder. LRRK2 is expressed by both neurons and microglia, the residential immune cells in the brain. Increasing evidence supports a role of LRRK2 in modulating microglial activity, of which Lrrk2-null rodent microglia display less inflammatory response to endotoxin lipopolysaccharide (LPS). The underlying molecular mechanism, however, remains elusive. Chemokine (C-X3-C) receptor 1 (CX3CR1), predominantly expressed by microglia, suppresses microglial inflammation while promotes migration. Using whole-genome microarray screening, we found that Cx3cr1 mRNA levels were substantially higher in microglia derived from Lrrk2 knockout (Lrrk2-/-) mice. The total and cell surface levels of CX3CR1 proteins were also remarkably increased. In correlation with the enhanced CX3CR1 expression, Lrrk2-null microglia migrated faster and travelled longer distance toward the source of fractalkine (CX3CL1), an endogenous ligand of CX3CR1. To investigate the impact of CX3CR1 elevation in vivo, we compared LPS-induced inflammation in the striatum of Lrrk2-/- knockout mice with Cx3cr1 heterozygous and homozygous knockout background. We found that a complete loss of Cx3cr1 restored the responsiveness of Lrrk2-/- microglia to LPS stimulation. In conclusion, our findings reveal a previously unknown regulatory role for LRRK2 in CX3CR1 signalling and suggest that an increase of CX3CR1 activity contributes to the attenuated inflammatory responses in Lrrk2-null microglia.
Collapse
Affiliation(s)
- Bo Ma
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Leyan Xu
- Division of Neuropathology, Department of pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaodong Pan
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Lixin Sun
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jinhui Ding
- Bioinformatics Core, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Chengsong Xie
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Vassilis E Koliatsos
- Division of Neuropathology, Department of pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huaibin Cai
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
115
|
Abstract
Lysosomes have emerged in the last decade as an immensely important intracellular site of Ca2+ storage and signalling. More recently there has been an increase in the number of new ion channels found to be functional on lysosomes and the potential roles that these signalling pathways might play in fundamental cellular processes are being uncovered. Defects in lysosomal function have been shown to result in changes in lysosomal Ca2+ homeostasis and ultimately can result in cell death. Several neurodegenerative diseases, from rare lysosomal storage diseases through to more common diseases of ageing, have recently been identified as having alterations in lysosomal Ca2+ homeostasis that may play an important role in neuronal excitotoxicity and ultimately cell death. This review will critically summarise these recent findings.
Collapse
Affiliation(s)
- Emyr Lloyd-Evans
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Museum Avenue, Cardiff, CF10 3AX
| |
Collapse
|
116
|
Activation Mechanism of LRRK2 and Its Cellular Functions in Parkinson's Disease. PARKINSONS DISEASE 2016; 2016:7351985. [PMID: 27293958 PMCID: PMC4880697 DOI: 10.1155/2016/7351985] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/19/2016] [Indexed: 01/09/2023]
Abstract
Human LRRK2 (Leucine-Rich Repeat Kinase 2) has been associated with both familial and idiopathic Parkinson's disease (PD). Although several LRRK2 mediated pathways and interaction partners have been identified, the cellular functions of LRRK2 and LRRK2 mediated progression of PD are still only partially understood. LRRK2 belongs to the group of Roco proteins which are characterized by the presence of a Ras-like G-domain (Roc), a C-terminal of Roc domain (COR), a kinase, and several protein-protein interaction domains. Roco proteins exhibit a complex activation mechanism involving intramolecular signaling, dimerization, and substrate/effector binding. Importantly, PD mutations in LRRK2 have been linked to a decreased GTPase and impaired kinase activity, thus providing putative therapeutic targets. To fully explore these potential targets it will be crucial to understand the function and identify the pathways responsible for LRRK2-linked PD. Here, we review the recent progress in elucidating the complex LRRK2 activation mechanism, describe the accumulating evidence that link LRRK2-mediated PD to mitochondrial dysfunction and aberrant autophagy, and discuss possible ways for therapeutically targeting LRRK2.
Collapse
|
117
|
Abstract
Mutations in LRRK2 are associated with inherited Parkinson's disease (PD) in a large number of families, and the genetic locus containing the LRRK2 gene contains a risk factor for sporadic PD. The LRRK2 protein contains several domains that suggest a role in cellular signaling, including a kinase domain. It is also clear that LRRK2 interacts, either physically or genetically, with several other important proteins implicated in PD, suggesting that LRRK2 may be a central player in the pathways that underlie parkinsonism. As such, LRRK2 has been proposed to be a plausible target for therapeutic intervention, with kinase inhibition being pursued most actively. However, there are still several fundamental aspects of LRRK2 biology and function that remain unresolved at this time. This review will focus on the key questions of normal function of LRRK2 and how this might be related to the pathophysiology of PD.
Collapse
|
118
|
Wang J, Song W. Regulation of LRRK2 promoter activity and gene expression by Sp1. Mol Brain 2016; 9:33. [PMID: 27004687 PMCID: PMC4802577 DOI: 10.1186/s13041-016-0215-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/14/2016] [Indexed: 12/12/2022] Open
Abstract
Background The dopaminergic neurodegeneration in the nigrostriatal pathway is a prominent neuropathological feature of Parkinson’s disease (PD). Mutations in various genes have been linked to familial PD, and leucine-rich repeat kinase 2 (LRRK2) gene is one of them. LRRK2 is a large complex protein, belonging to the ROCO family of proteins. Recent studies suggest that the level of LRRK2 protein is one of the contributing factors to PD pathogenesis. However, it remains elusive how LRRK2 is regulated at the transcriptional and translational level. Results In this study, we cloned a 1738 bp 5’-flanking region of the human LRRK2 gene. The transcriptional start site (TSS) was located to 135 bp upstream of translational start site and the fragment −118 to +133 bp had the minimum promoter activity required for transcription. There were two functional Sp1- responsive elements on the human LRRK2 gene promoter revealed by electrophoretic mobility shift assay (EMSA). Sp1 overexpression promoted LRRK2 transcription and translation in the cellular model. On the contrary, application of mithramycin A inhibited LRRK2 transcriptional and translational activities. Conclusion This is the first study indicating that Sp1 signaling plays an important role in the regulation of human LRRK2 gene expression. It suggests that controlling LRRK2 level by manipulating Sp1 signaling may be beneficial to attenuate PD-related neuropathology.
Collapse
Affiliation(s)
- Juelu Wang
- Department of Psychiatry, Townsend Family Laboratories, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Department of Psychiatry, Townsend Family Laboratories, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
119
|
Bae JR, Lee BD. Function and dysfunction of leucine-rich repeat kinase 2 (LRRK2): Parkinson's disease and beyond. BMB Rep 2016; 48:243-8. [PMID: 25703537 PMCID: PMC4578562 DOI: 10.5483/bmbrep.2015.48.5.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Indexed: 12/19/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of
familial Parkinson’s disease (PD). As such, functions and dysfunctions of LRRK2
in PD have been the subject of extensive investigation. In addition to PD,
increasing evidence is suggesting that LRRK2 is associated with a wide range of
diseases. Genome-wide association studies have implicated LRRK2 in Crohn’s
disease (CD) and leprosy, and the carriers with pathogenic mutations of LRRK2
show increased risk to develop particular types of cancer. LRRK2 mutations are
rarely found in Alzheimer’s disease (AD), but LRRK2 might play a part in
tauopathies. The association of LRRK2 with the pathogenesis of apparently
unrelated diseases remains enigmatic, but it might be related to the yet unknown
diverse functions of LRRK2. Here, we reviewed current knowledge on the link
between LRRK2 and several diseases, including PD, AD, CD, leprosy, and cancer,
and discussed the possibility of targeting LRRK2 in such diseases. [BMB Reports
2015; 48(5): 243-248]
Collapse
Affiliation(s)
- Jae Ryul Bae
- Department of Neuroscience, of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Byoung Dae Lee
- Department of Neuroscience; Neurodegeneration Control Research Center; Department of Physiology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
120
|
Alterations in late endocytic trafficking related to the pathobiology of LRRK2-linked Parkinson's disease. Biochem Soc Trans 2016; 43:390-5. [PMID: 26009181 DOI: 10.1042/bst20140301] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene comprise the most common cause of familial Parkinson's disease (PD), and variants increase the risk for sporadic PD. LRRK2 displays kinase and GTPase activity, and altered catalytic activity correlates with neurotoxicity, making LRRK2 a promising therapeutic target. Despite the importance of LRRK2 for disease pathogenesis, its normal cellular function, and the mechanism(s) by which pathogenic mutations cause neurodegeneration remain unclear. LRRK2 seems to regulate a variety of intracellular vesicular trafficking events to and from the late endosome in a manner dependent on various Rab proteins. At least some of those events are further regulated by LRRK2 in a manner dependent on two-pore channels (TPCs). TPCs are ionic channels localized to distinct endosomal structures and can cause localized calcium release from those acidic stores, with downstream effects on vesicular trafficking. Here, we review current knowledge about the link between LRRK2, TPC- and Rab-mediated vesicular trafficking to and from the late endosome, highlighting a possible cross-talk between endolysosomal calcium stores and Rab proteins underlying pathomechanism(s) in LRRK2-related PD.
Collapse
|
121
|
Two-pore channels at the intersection of endolysosomal membrane traffic. Biochem Soc Trans 2016; 43:434-41. [PMID: 26009187 DOI: 10.1042/bst20140303] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Two-pore channels (TPCs) are ancient members of the voltage-gated ion channel superfamily that localize to acidic organelles such as lysosomes. The TPC complex is the proposed target of the Ca2+-mobilizing messenger NAADP, which releases Ca2+ from these acidic Ca2+ stores. Whereas details of TPC activation and native ion permeation remain unclear, a consensus has emerged around their function in regulating endolysosomal trafficking. This role is supported by recent proteomic data showing that TPCs interact with proteins controlling membrane organization and dynamics, including Rab GTPases and components of the fusion apparatus. Regulation of TPCs by PtdIns(3,5)P2 and/or NAADP (nicotinic acid adenine dinucleotide phosphate) together with their functional and physical association with Rab proteins provides a mechanism for coupling phosphoinositide and trafficking protein cues to local ion fluxes. Therefore, TPCs work at the regulatory cross-roads of (patho)physiological cues to co-ordinate and potentially deregulate traffic flow through the endolysosomal network. This review focuses on the native role of TPCs in trafficking and their emerging contributions to endolysosomal trafficking dysfunction.
Collapse
|
122
|
Langston RG, Rudenko IN, Cookson MR. The function of orthologues of the human Parkinson's disease gene LRRK2 across species: implications for disease modelling in preclinical research. Biochem J 2016; 473:221-32. [PMID: 26811536 PMCID: PMC5165698 DOI: 10.1042/bj20150985] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the period since LRRK2 (leucine-rich repeat kinase 2) was identified as a causal gene for late-onset autosomal dominant parkinsonism, a great deal of work has been aimed at understanding whether the LRRK2 protein might be a druggable target for Parkinson's disease (PD). As part of this effort, animal models have been developed to explore both the normal and the pathophysiological roles of LRRK2. However, LRRK2 is part of a wider family of proteins whose functions in different organisms remain poorly understood. In this review, we compare the information available on biochemical properties of LRRK2 homologues and orthologues from different species from invertebrates (e.g. Caenorhabditis elegans and Drosophila melanogaster) to mammals. We particularly discuss the mammalian LRRK2 homologue, LRRK1, and those species where there is only a single LRRK homologue, discussing examples where each of the LRRK family of proteins has distinct properties as well as those cases where there appear to be functional redundancy. We conclude that uncovering the function of LRRK2 orthologues will help to elucidate the key properties of human LRRK2 as well as to improve understanding of the suitability of different animal models for investigation of LRRK2-related PD.
Collapse
Affiliation(s)
- Rebekah G. Langston
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Iakov N. Rudenko
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| |
Collapse
|
123
|
Kilpatrick BS, Magalhaes J, Beavan MS, McNeill A, Gegg ME, Cleeter MWJ, Bloor-Young D, Churchill GC, Duchen MR, Schapira AH, Patel S. Endoplasmic reticulum and lysosomal Ca²⁺ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts. Cell Calcium 2015; 59:12-20. [PMID: 26691915 PMCID: PMC4751977 DOI: 10.1016/j.ceca.2015.11.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/06/2015] [Accepted: 11/25/2015] [Indexed: 11/19/2022]
Abstract
Mutations in β-glucocerebrosidase (encoded by GBA1) cause Gaucher disease (GD), a lysosomal storage disorder, and increase the risk of developing Parkinson disease (PD). The pathogenetic relationship between the two disorders is unclear. Here, we characterised Ca(2+) release in fibroblasts from type I GD and PD patients together with age-matched, asymptomatic carriers, all with the common N370S mutation in β-glucocerebrosidase. We show that endoplasmic reticulum (ER) Ca(2+) release was potentiated in GD and PD patient fibroblasts but not in cells from asymptomatic carriers. ER Ca(2+) signalling was also potentiated in fibroblasts from aged healthy subjects relative to younger individuals but not further increased in aged PD patient cells. Chemical or molecular inhibition of β-glucocerebrosidase in fibroblasts and a neuronal cell line did not affect ER Ca(2+) signalling suggesting defects are independent of enzymatic activity loss. Conversely, lysosomal Ca(2+) store content was reduced in PD fibroblasts and associated with age-dependent alterations in lysosomal morphology. Accelerated remodelling of Ca(2+) stores by pathogenic GBA1 mutations may therefore feature in PD.
Collapse
Affiliation(s)
- Bethan S Kilpatrick
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Joana Magalhaes
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Michelle S Beavan
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Alisdair McNeill
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Matthew E Gegg
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Michael W J Cleeter
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | | | - Grant C Churchill
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anthony H Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
124
|
Rodriguez-Fernandez IA, Dell’Angelica EC. Identification of Atg2 and ArfGAP1 as Candidate Genetic Modifiers of the Eye Pigmentation Phenotype of Adaptor Protein-3 (AP-3) Mutants in Drosophila melanogaster. PLoS One 2015; 10:e0143026. [PMID: 26565960 PMCID: PMC4643998 DOI: 10.1371/journal.pone.0143026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/29/2015] [Indexed: 11/19/2022] Open
Abstract
The Adaptor Protein (AP)-3 complex is an evolutionary conserved, molecular sorting device that mediates the intracellular trafficking of proteins to lysosomes and related organelles. Genetic defects in AP-3 subunits lead to impaired biogenesis of lysosome-related organelles (LROs) such as mammalian melanosomes and insect eye pigment granules. In this work, we have performed a forward screening for genetic modifiers of AP-3 function in the fruit fly, Drosophila melanogaster. Specifically, we have tested collections of large multi-gene deletions–which together covered most of the autosomal chromosomes–to identify chromosomal regions that, when deleted in single copy, enhanced or ameliorated the eye pigmentation phenotype of two independent AP-3 subunit mutants. Fine-mapping led us to define two non-overlapping, relatively small critical regions within fly chromosome 3. The first critical region included the Atg2 gene, which encodes a conserved protein involved in autophagy. Loss of one functional copy of Atg2 ameliorated the pigmentation defects of mutants in AP-3 subunits as well as in two other genes previously implicated in LRO biogenesis, namely Blos1 and lightoid, and even increased the eye pigment content of wild-type flies. The second critical region included the ArfGAP1 gene, which encodes a conserved GTPase-activating protein with specificity towards GTPases of the Arf family. Loss of a single functional copy of the ArfGAP1 gene ameliorated the pigmentation phenotype of AP-3 mutants but did not to modify the eye pigmentation of wild-type flies or mutants in Blos1 or lightoid. Strikingly, loss of the second functional copy of the gene did not modify the phenotype of AP-3 mutants any further but elicited early lethality in males and abnormal eye morphology when combined with mutations in Blos1 and lightoid, respectively. These results provide genetic evidence for new functional links connecting the machinery for biogenesis of LROs with molecules implicated in autophagy and small GTPase regulation.
Collapse
Affiliation(s)
- Imilce A. Rodriguez-Fernandez
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Esteban C. Dell’Angelica
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
125
|
Imai Y, Kobayashi Y, Inoshita T, Meng H, Arano T, Uemura K, Asano T, Yoshimi K, Zhang CL, Matsumoto G, Ohtsuka T, Kageyama R, Kiyonari H, Shioi G, Nukina N, Hattori N, Takahashi R. The Parkinson's Disease-Associated Protein Kinase LRRK2 Modulates Notch Signaling through the Endosomal Pathway. PLoS Genet 2015; 11:e1005503. [PMID: 26355680 PMCID: PMC4565672 DOI: 10.1371/journal.pgen.1005503] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/14/2015] [Indexed: 12/03/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of familial and idiopathic Parkinson’s disease (PD). We have identified two novel LRRK2-associated proteins, a HECT-type ubiquitin ligase, HERC2, and an adaptor-like protein with six repeated Neuralized domains, NEURL4. LRRK2 binds to NEURL4 and HERC2 via the LRRK2 Ras of complex proteins (ROC) domain and NEURL4, respectively. HERC2 and NEURL4 link LRRK2 to the cellular vesicle transport pathway and Notch signaling, through which the LRRK2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. This process negatively regulates Notch signaling through cis-inhibition by stabilizing Dll1/Dl, which accelerates neural stem cell differentiation and modulates the function and survival of differentiated dopaminergic neurons. These effects are strengthened by the R1441G ROC domain-mutant of LRRK2. These findings suggest that the alteration of Notch signaling in mature neurons is a component of PD etiology linked to LRRK2. LRRK2 is linked to autosomal dominant late-onset Parkinson’s disease, suggesting that LRRK2 gain-of-function mutations lead to age-dependent degeneration of the midbrain dopaminergic neurons. In this study, we describe two novel LRRK2-associated proteins HERC2 and NEURL4, which are a ubiquitin ligase and an adaptor-like protein, respectively. HERC2 and NEURL4 direct LRRK2 to Notch signaling pathway, in which the LRRK2-NEURL4-HERC2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. As a result, the amounts of Dll1/D1 on the plasma membrane are increased, which affects negatively Notch signaling through cis-inhibition. The effect is enhanced by a Parkinson’s-disease associated mutation of LRRK2. Inhibition of Notch signaling in adult dopaminergic neurons impairs its functions and survival. These findings indicate a possible link between Notch pathway and Parkinson’s disease.
Collapse
Affiliation(s)
- Yuzuru Imai
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- * E-mail: (YI); (NH); (RT)
| | - Yoshito Kobayashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
| | - Tsuyoshi Inoshita
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hongrui Meng
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Taku Arano
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kengo Uemura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Asano
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Yoshimi
- Department of Neurophysiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chang-Liang Zhang
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
| | - Gen Matsumoto
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiyuki Ohtsuka
- Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Go Shioi
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Nobuyuki Nukina
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- * E-mail: (YI); (NH); (RT)
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
- * E-mail: (YI); (NH); (RT)
| |
Collapse
|
126
|
Beilina A, Cookson MR. Genes associated with Parkinson's disease: regulation of autophagy and beyond. J Neurochem 2015. [PMID: 26223426 DOI: 10.1111/jnc.13266] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial progress has been made in the genetic basis of Parkinson's disease (PD). In particular, by identifying genes that segregate with inherited PD or show robust association with sporadic disease, and by showing the same genes are found on both lists, we have generated an outline of the cause of this condition. Here, we will discuss what those genes tell us about the underlying biology of PD. We specifically discuss the relationships between protein products of PD genes and show that common links include regulation of the autophagy-lysosome system, an important way by which cells recycle proteins and organelles. We also discuss whether all PD genes should be considered to be in the same pathway and propose that in some cases the relationships are closer, whereas in other cases the interactions are more distant and might be considered separate. Beilina and Cookson review the links between genes for Parkinson's disease (red) and the autophagy-lysosomal system. They propose the hypothesis that many of the known PD genes can be assigned to pathways that affect (I) turnover of mitochondria via mitophagy (II) turnover of several vesicular structures via macroautophagy or chaperone-mediated autophagy or (III) general lysosome function. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Alexandra Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.
| |
Collapse
|
127
|
Henry AG, Aghamohammadzadeh S, Samaroo H, Chen Y, Mou K, Needle E, Hirst WD. Pathogenic LRRK2 mutations, through increased kinase activity, produce enlarged lysosomes with reduced degradative capacity and increase ATP13A2 expression. Hum Mol Genet 2015; 24:6013-28. [PMID: 26251043 DOI: 10.1093/hmg/ddv314] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/29/2015] [Indexed: 12/12/2022] Open
Abstract
Lysosomal dysfunction plays a central role in the pathogenesis of several neurodegenerative disorders, including Parkinson's disease (PD). Several genes linked to genetic forms of PD, including leucine-rich repeat kinase 2 (LRRK2), functionally converge on the lysosomal system. While mutations in LRRK2 are commonly associated with autosomal-dominant PD, the physiological and pathological functions of this kinase remain poorly understood. Here, we demonstrate that LRRK2 regulates lysosome size, number and function in astrocytes, which endogenously express high levels of LRRK2. Expression of LRRK2 G2019S, the most common pathological mutation, produces enlarged lysosomes and diminishes the lysosomal capacity of these cells. Enlarged lysosomes appears to be a common phenotype associated with pathogenic LRRK2 mutations, as we also observed this effect in cells expressing other LRRK2 mutations; R1441C or Y1699C. The lysosomal defects associated with these mutations are dependent on both the catalytic activity of the kinase and autophosphorylation of LRRK2 at serine 1292. Further, we demonstrate that blocking LRRK2's kinase activity, with the potent and selective inhibitor PF-06447475, rescues the observed defects in lysosomal morphology and function. The present study also establishes that G2019S mutation leads to a reduction in lysosomal pH and increased expression of the lysosomal ATPase ATP13A2, a gene linked to a parkinsonian syndrome (Kufor-Rakeb syndrome), in brain samples from mouse and human LRRK2 G2019S carriers. Together, these results demonstrate that PD-associated LRRK2 mutations perturb lysosome function in a kinase-dependent manner, highlighting the therapeutic promise of LRRK2 kinase inhibitors in the treatment of PD.
Collapse
Affiliation(s)
- Anastasia G Henry
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Soheil Aghamohammadzadeh
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Harry Samaroo
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Yi Chen
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Kewa Mou
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Elie Needle
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Warren D Hirst
- Pfizer Neuroscience and Pain Research Unit, Pfizer Global Research and Development, Cambridge, MA 02139, USA
| |
Collapse
|
128
|
Lin CH, Li H, Lee YN, Cheng YJ, Wu RM, Chien CT. Lrrk regulates the dynamic profile of dendritic Golgi outposts through the golgin Lava lamp. J Cell Biol 2015. [PMID: 26216903 PMCID: PMC4523617 DOI: 10.1083/jcb.201411033] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lrrk regulates Golgi outpost (GOP) dynamics in dendrites by antagonizing the interaction between the golgin Lva and dynein heavy chain at GOPs, thereby disrupting minus end–directed transport along dendritic microtubules by dynein. Constructing the dendritic arbor of neurons requires dynamic movements of Golgi outposts (GOPs), the prominent component in the dendritic secretory pathway. GOPs move toward dendritic ends (anterograde) or cell bodies (retrograde), whereas most of them remain stationary. Here, we show that Leucine-rich repeat kinase (Lrrk), the Drosophila melanogaster homologue of Parkinson’s disease–associated Lrrk2, regulates GOP dynamics in dendrites. Lrrk localized at stationary GOPs in dendrites and suppressed GOP movement. In Lrrk loss-of-function mutants, anterograde movement of GOPs was enhanced, whereas Lrrk overexpression increased the pool size of stationary GOPs. Lrrk interacted with the golgin Lava lamp and inhibited the interaction between Lva and dynein heavy chain, thus disrupting the recruitment of dynein to Golgi membranes. Whereas overexpression of kinase-dead Lrrk caused dominant-negative effects on GOP dynamics, overexpression of the human LRRK2 mutant G2019S with augmented kinase activity promoted retrograde movement. Our study reveals a pathogenic pathway for LRRK2 mutations causing dendrite degeneration.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hsun Li
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 115, Taiwan
| | - Yi-Nan Lee
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ying-Ju Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
129
|
Liu G, Sgobio C, Gu X, Sun L, Lin X, Yu J, Parisiadou L, Xie C, Sastry N, Ding J, Lohr KM, Miller GW, Mateo Y, Lovinger DM, Cai H. Selective expression of Parkinson's disease-related Leucine-rich repeat kinase 2 G2019S missense mutation in midbrain dopaminergic neurons impairs dopamine release and dopaminergic gene expression. Hum Mol Genet 2015; 24:5299-312. [PMID: 26123485 DOI: 10.1093/hmg/ddv249] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/23/2015] [Indexed: 01/25/2023] Open
Abstract
Preferential dysfunction/degeneration of midbrain substantia nigra pars compacta (SNpc) dopaminergic (DA) neurons contributes to the main movement symptoms manifested in Parkinson's disease (PD). Although the Leucine-rich repeat kinase 2 (LRRK2) G2019S missense mutation (LRRK2 G2019S) is the most common causative genetic factor linked to PD, the effects of LRRK2 G2019S on the function and survival of SNpc DA neurons are poorly understood. Using a binary gene expression system, we generated transgenic mice expressing either wild-type human LRRK2 (WT mice) or the LRRK2 G2019S mutation (G2019S mice) selectively in the midbrain DA neurons. Here we show that overexpression of LRRK2 G2019S did not induce overt motor abnormalities or substantial SNpc DA neuron loss. However, the LRRK2 G2019S mutation impaired dopamine homeostasis and release in aged mice. This reduction in dopamine content/release coincided with the degeneration of DA axon terminals and decreased expression of DA neuron-enriched genes tyrosine hydroxylase (TH), vesicular monoamine transporter 2, dopamine transporter and aldehyde dehydrogenase 1. These factors are responsible for dopamine synthesis, transport and degradation, and their expression is regulated by transcription factor paired-like homeodomain 3 (PITX3). Levels of Pitx3 mRNA and protein were similarly decreased in the SNpc DA neurons of aged G2019S mice. Together, these findings suggest that PITX3-dependent transcription regulation could be one of the many potential mechanisms by which LRRK2 G2019S acts in SNpc DA neurons, resulting in downregulation of its downstream target genes critical for dopamine homeostasis and release.
Collapse
Affiliation(s)
- Guoxiang Liu
- Transgenics Section and Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | - Loukia Parisiadou
- Transgenics Section and Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | - Namratha Sastry
- Transgenics Section and Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jinhui Ding
- Bioinformatics Core, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly M Lohr
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA and
| | - Gary W Miller
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA and
| | - Yolanda Mateo
- Laboratory of Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20852, USA
| | - David M Lovinger
- Laboratory of Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20852, USA
| | | |
Collapse
|
130
|
Leucine-Rich Repeat Kinase 1 Regulates Autophagy through Turning On TBC1D2-Dependent Rab7 Inactivation. Mol Cell Biol 2015; 35:3044-58. [PMID: 26100023 DOI: 10.1128/mcb.00085-15] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/19/2015] [Indexed: 01/23/2023] Open
Abstract
Autophagy is a conserved process that enables catabolic and degradative pathways. Rab family proteins, which are active in the GTP-bound form, regulate the transport and fusion of autophagosomes. However, it remains unclear how each cycle of Rab activation and inactivation is precisely regulated. Here, we show that leucine-rich repeat kinase 1 (LRRK1) regulates autophagic flux by controlling Rab7 activity in autolysosome formation. Upon induction of autophagy, LRRK1 was recruited via an association with VAMP7 to the autolysosome, where it activated the Rab7 GTPase-activating protein (GAP) TBC1D2, thereby switching off Rab7 signaling. Consistent with this model, LRRK1 deletion caused mice to be vulnerable to starvation and disrupted autolysosome formation, as evidenced by the accumulation of enlarged autolysosomes with undegraded LC3-II and persistently high levels of Rab7-GTP. This defect in autophagic flux was partially rescued by a mutant form of TBC1D2 with elevated Rab7-GAP activity. Thus, the spatiotemporal regulation of Rab7 activity during tunicamycin-induced autophagy is regulated by LRRK1.
Collapse
|
131
|
De Rosa P, Marini ES, Gelmetti V, Valente EM. Candidate genes for Parkinson disease: Lessons from pathogenesis. Clin Chim Acta 2015; 449:68-76. [PMID: 26048192 DOI: 10.1016/j.cca.2015.04.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 01/06/2023]
Abstract
Parkinson disease (PD) is a multifactorial neurodegenerative disease characterized by the progressive loss of specific neuronal populations and accumulation of Lewy bodies in the brain, leading to motor and non-motor symptoms. In a small subset of patients, PD is dominantly or recessively inherited, while a number of susceptibility genetic loci have been identified through genome wide association studies. The discovery of genes mutated in PD and functional studies on their protein products have provided new insights into the molecular events leading to neurodegeneration, suggesting that few interconnected molecular pathways may be deranged in all forms of PD, triggering neuronal loss. Here, we summarize the most relevant findings implicating the main PD-related proteins in biological processes such as mitochondrial dysfunction, misfolded protein damage, alteration of cellular clearance systems, abnormal calcium handling and altered inflammatory response, which represent key targets for neuroprotection.
Collapse
Affiliation(s)
- Priscilla De Rosa
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy
| | - Elettra Sara Marini
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy; Dept. of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Vania Gelmetti
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy
| | - Enza Maria Valente
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy; Section of Neurosciences, Dept. of Medicine and Surgery, University of Salerno, Salerno, Italy.
| |
Collapse
|
132
|
Vanhauwaert R, Verstreken P. Flies with Parkinson's disease. Exp Neurol 2015; 274:42-51. [PMID: 25708988 DOI: 10.1016/j.expneurol.2015.02.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 02/11/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is an incurable neurodegenerative disease. Most cases of the disease are of sporadic origin, but about 10% of the cases are familial. The genes thus far identified in Parkinson's disease are well conserved. Drosophila is ideally suited to study the molecular neuronal cell biology of these genes and the pathogenic mutations in Parkinson's disease. Flies reproduce quickly, and their elaborate genetic tools in combination with their small size allow researchers to analyze identified cells and neurons in large numbers of animals. Furthermore, fruit flies recapitulate many of the cellular and molecular defects also seen in patients, and these defects often result in clear locomotor and behavioral phenotypes, facilitating genetic modifier screens. Hence, Drosophila has played a prominent role in Parkinson's disease research and has provided invaluable insight into the molecular mechanisms of this disease.
Collapse
Affiliation(s)
- Roeland Vanhauwaert
- VIB Center for the Biology of Disease, KU Leuven, Herestraat 49,3000 Leuven, Belgium; Laboratory of Neuronal Communication, Leuven Institute for Neurodegenerative Disease (LIND), Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Patrik Verstreken
- VIB Center for the Biology of Disease, KU Leuven, Herestraat 49,3000 Leuven, Belgium; Laboratory of Neuronal Communication, Leuven Institute for Neurodegenerative Disease (LIND), Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
133
|
Perrett RM, Alexopoulou Z, Tofaris GK. The endosomal pathway in Parkinson's disease. Mol Cell Neurosci 2015; 66:21-8. [PMID: 25701813 DOI: 10.1016/j.mcn.2015.02.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/22/2023] Open
Abstract
Parkinson's disease is primarily a movement disorder with predilection for the nigral dopaminergic neurons and is often associated with widespread neurodegeneration and diffuse Lewy body deposition. Recent advances in molecular genetics and studies in model organisms have transformed our understanding of Parkinson's pathogenesis and suggested unifying biochemical pathways despite the clinical heterogeneity of the disease. In this review, we summarized the evidence that a number of Parkinson's associated genetic mutations or polymorphisms (LRRK2, VPS35, GBA, ATP13A2, ATP6AP2, DNAJC13/RME-8, RAB7L1, GAK) disrupt protein trafficking and degradation via the endosomal pathway and discussed how such defects could arise from or contribute to the accumulation and misfolding of α-synuclein in Lewy bodies. We propose that an age-related pathological depletion of functional endolysosomes due to neuromelanin deposition in dopaminergic neurons may increase their susceptibility to stochastic molecular defects in this pathway and we discuss how enzymes that regulate ubiquitin signaling, as exemplified by the ubiquitin ligase Nedd4, could provide the missing link between genetic and acquired defects in endosomal trafficking. This article is part of a Special Issue entitled 'Neuronal Protein'.
Collapse
Affiliation(s)
- Rebecca M Perrett
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Zoi Alexopoulou
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK.
| |
Collapse
|
134
|
Daniel G, Moore DJ. Modeling LRRK2 Pathobiology in Parkinson's Disease: From Yeast to Rodents. Curr Top Behav Neurosci 2015; 22:331-368. [PMID: 24850078 DOI: 10.1007/7854_2014_311] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2, PARK8) gene represent the most common cause of familial Parkinson's disease (PD) with autosomal dominant inheritance, whereas common variation at the LRRK2 genomic locus influences the risk of developing idiopathic PD. LRRK2 is a member of the ROCO protein family and contains multiple domains, including Ras-of-Complex (ROC) GTPase, kinase, and protein-protein interaction domains. In the last decade, the biochemical characterization of LRRK2 and the development of animal model s have provided important insight into the pathobiology of LRRK2. In this review, we comprehensively describe the different models employed to understand LRRK2-associated PD, including yeast, invertebrates, transgenic and viral-based rodents, and patient-derived induced pluripotent stem cells. We discuss how these models have contributed to understanding LRRK2 pathobiology and the advantages and limitations of each model for exploring aspects of LRRK2-associated PD.
Collapse
Affiliation(s)
- Guillaume Daniel
- School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | | |
Collapse
|
135
|
Phosphorylation of LRRK2 by casein kinase 1α regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat Commun 2014; 5:5827. [PMID: 25500533 PMCID: PMC4268884 DOI: 10.1038/ncomms6827] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/10/2014] [Indexed: 12/29/2022] Open
Abstract
LRRK2, a gene relevant to Parkinson's disease, encodes a scaffolding protein with both GTPase and kinase activities. LRRK2 protein is itself phosphorylated and therefore subject to regulation by cell signaling but the kinase(s) responsible for this event have not been definitively identified. Here, using an unbiased siRNA kinome screen, we identify and validate casein kinase 1α (CK1α) as being responsible for LRRK2 phosphorylation, including in the adult mouse striatum. We further show that LRRK2 recruitment to TGN46-positive Golgi-derived vesicles is modulated by constitutive LRRK2 phosphorylation by CK1α. These effects are mediated by differential protein interactions of LRRK2 with a guanine nucleotide exchange factor, ARHGEF7. These pathways are therefore likely involved in the physiological maintenance of the Golgi in cells, which may play a role in the pathogenesis of Parkinson's disease.
Collapse
|
136
|
Schreij AMA, Chaineau M, Ruan W, Lin S, Barker PA, Fon EA, McPherson PS. LRRK2 localizes to endosomes and interacts with clathrin-light chains to limit Rac1 activation. EMBO Rep 2014; 16:79-86. [PMID: 25427558 DOI: 10.15252/embr.201438714] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of dominant-inherited Parkinson's disease (PD), and yet we do not fully understand the physiological function(s) of LRRK2. Various components of the clathrin machinery have been recently found mutated in familial forms of PD. Here, we provide molecular insight into the association of LRRK2 with the clathrin machinery. We report that through its GTPase domain, LRRK2 binds directly to clathrin-light chains (CLCs). Using genome-edited HA-LRRK2 cells, we localize LRRK2 to endosomes on the degradative pathway, where it partially co-localizes with CLCs. Knockdown of CLCs and/or LRRK2 enhances the activation of the small GTPase Rac1, leading to alterations in cell morphology, including the disruption of neuronal dendritic spines. In Drosphila, a minimal rough eye phenotype caused by overexpression of Rac1, is dramatically enhanced by loss of function of CLC and LRRK2 homologues, confirming the importance of this pathway in vivo. Our data identify a new pathway in which CLCs function with LRRK2 to control Rac1 activation on endosomes, providing a new link between the clathrin machinery, the cytoskeleton and PD.
Collapse
Affiliation(s)
- Andrea M A Schreij
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Mathilde Chaineau
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Wenjing Ruan
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Susan Lin
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Philip A Barker
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| | - Peter S McPherson
- Department of Neurology and Neurosurgery and McGill Parkinson Program, Montreal Neurological Institute McGill University, Montreal, Quebec, Canada
| |
Collapse
|
137
|
Hockey LN, Kilpatrick BS, Eden ER, Lin-Moshier Y, Brailoiu GC, Brailoiu E, Futter CE, Schapira AH, Marchant JS, Patel S. Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition. J Cell Sci 2014; 128:232-8. [PMID: 25416817 PMCID: PMC4294771 DOI: 10.1242/jcs.164152] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Two-pore channels (TPCs) are endolysosomal ion channels implicated in Ca2+ signalling from acidic organelles. The relevance of these ubiquitous proteins for human disease, however, is unclear. Here, we report that lysosomes are enlarged and aggregated in fibroblasts from Parkinson disease patients with the common G2019S mutation in LRRK2. Defects were corrected by molecular silencing of TPC2, pharmacological inhibition of TPC regulators [Rab7, NAADP and PtdIns(3,5)P2] and buffering local Ca2+ increases. NAADP-evoked Ca2+ signals were exaggerated in diseased cells. TPC2 is thus a potential drug target within a pathogenic LRRK2 cascade that disrupts Ca2+-dependent trafficking in Parkinson disease.
Collapse
Affiliation(s)
- Leanne N Hockey
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Bethan S Kilpatrick
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Emily R Eden
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Yaping Lin-Moshier
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, 55455, USA
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University, Jefferson School of Pharmacy, Philadelphia, 19107, USA
| | - Eugen Brailoiu
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, 19140, USA
| | - Clare E Futter
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Anthony H Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Jonathan S Marchant
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, 55455, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
138
|
Esteves AR, G-Fernandes M, Santos D, Januário C, Cardoso SM. The Upshot of LRRK2 Inhibition to Parkinson’s Disease Paradigm. Mol Neurobiol 2014; 52:1804-1820. [DOI: 10.1007/s12035-014-8980-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/31/2014] [Indexed: 12/13/2022]
|
139
|
Li JQ, Tan L, Yu JT. The role of the LRRK2 gene in Parkinsonism. Mol Neurodegener 2014; 9:47. [PMID: 25391693 PMCID: PMC4246469 DOI: 10.1186/1750-1326-9-47] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/21/2014] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD), like many common age-related conditions, has been recognized to have a substantial genetic component. Multiple lines of evidence suggest that Leucine-rich repeat kinase 2 (LRRK2) is a crucial factor to understanding the etiology of PD. LRRK2 is a large, widely expressed, multi-domain and multifunctional protein. LRRK2 mutations are the major cause to inherited and sporadic PD. In this review, we discuss the pathology and clinical features which show diversity and variability of LRRK2-associated PD. In addition, we do a thorough literature review and provide theoretical data for gene counseling. Further, we present the evidence linking LRRK2 to various possible pathogenic mechanism of PD such as α-synuclein, tau, inflammatory response, oxidative stress, mitochondrial dysfunction, synaptic dysfunction as well as autophagy-lysosomal system. Based on the above work, we investigate activities both within GTPase and outside enzymatic regions in order to obtain a potential therapeutic approach to solve the LRRK2 problem.
Collapse
Affiliation(s)
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No, 5 Donghai Middle Road, Qingdao 266071, PR China.
| | | |
Collapse
|
140
|
Toward a comprehensive map of the effectors of rab GTPases. Dev Cell 2014; 31:358-373. [PMID: 25453831 PMCID: PMC4232348 DOI: 10.1016/j.devcel.2014.10.007] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/25/2014] [Accepted: 09/25/2014] [Indexed: 11/24/2022]
Abstract
The Rab GTPases recruit peripheral membrane proteins to intracellular organelles. These Rab effectors typically mediate the motility of organelles and vesicles and contribute to the specificity of membrane traffic. However, for many Rabs, few, if any, effectors have been identified; hence, their role remains unclear. To identify Rab effectors, we used a comprehensive set of Drosophila Rabs for affinity chromatography followed by mass spectrometry to identify the proteins bound to each Rab. For many Rabs, this revealed specific interactions with Drosophila orthologs of known effectors. In addition, we found numerous Rab-specific interactions with known components of membrane traffic as well as with diverse proteins not previously linked to organelles or having no known function. We confirm over 25 interactions for Rab2, Rab4, Rab5, Rab6, Rab7, Rab9, Rab18, Rab19, Rab30, and Rab39. These include tethering complexes, coiled-coiled proteins, motor linkers, Rab regulators, and several proteins linked to human disease. Proteomic screen identifies effectors of Drosophila Rabs with a human ortholog Specific hits include orthologs of numerous known effectors of mammalian Rabs Validated effectors include traffic proteins and those of unknown function Orthologs of disease genes CLEC16A, LRRK2, and SPG20 are validated as effectors
Collapse
|
141
|
Waschbüsch D, Michels H, Strassheim S, Ossendorf E, Kessler D, Gloeckner CJ, Barnekow A. LRRK2 transport is regulated by its novel interacting partner Rab32. PLoS One 2014; 9:e111632. [PMID: 25360523 PMCID: PMC4216093 DOI: 10.1371/journal.pone.0111632] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 09/29/2014] [Indexed: 11/18/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a multi-domain 280 kDa protein that is linked to Parkinson's disease (PD). Mutations especially in the GTPase and kinase domains of LRRK2 are the most common causes of heritable PD and are also found in sporadic forms of PD. Although the cellular function of LRRK2 is largely unknown there is increasing evidence that these mutations cause cell death due to autophagic dysfunction and mitochondrial damage. Here, we demonstrate a novel mechanism of LRRK2 binding and transport, which involves the small GTPases Rab32 and Rab38. Rab32 and its closest homologue Rab38 are known to organize the trans-Golgi network and transport of key enzymes in melanogenesis, whereas their function in non-melanogenic cells is still not well understood. Cellular processes such as autophagy, mitochondrial dynamics, phagocytosis or inflammatory processes in the brain have previously been linked to Rab32. Here, we demonstrate that Rab32 and Rab38, but no other GTPase tested, directly interact with LRRK2. GFP-Trap analyses confirmed the interaction of Rab32 with the endogenous LRRK2. In yeast two-hybrid experiments we identified a predicted coiled-coil motif containing region within the aminoterminus of LRRK2 as the possible interacting domain. Fluorescence microscopy demonstrated a co-localization of Rab32 and LRRK2 at recycling endosomes and transport vesicles, while overexpression of a constitutively active mutant of Rab32 led to an increased co-localization with Rab7/9 positive perinuclear late endosomes/MVBs. Subcellular fractionation experiments supported the novel role of Rab32 in LRRK2 late endosomal transport and sorting in the cell. Thus, Rab32 may regulate the physiological functions of LRRK2.
Collapse
Affiliation(s)
- Dieter Waschbüsch
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
- * E-mail:
| | - Helen Michels
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Swantje Strassheim
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Edith Ossendorf
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Daniel Kessler
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Christian Johannes Gloeckner
- Research Unit Protein Science, Helmholtz Zentrum München, Neuherberg, Germany
- Medical Proteome Center, Institute for Ophthalmic Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Angelika Barnekow
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| |
Collapse
|
142
|
Martin I, Kim JW, Dawson VL, Dawson TM. LRRK2 pathobiology in Parkinson's disease. J Neurochem 2014; 131:554-65. [PMID: 25251388 DOI: 10.1111/jnc.12949] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/14/2014] [Accepted: 09/16/2014] [Indexed: 12/11/2022]
Abstract
Mutations in the catalytic Roc-COR and kinase domains of leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial Parkinson's disease (PD). LRRK2 mutations cause PD with age-related penetrance and clinical features identical to late-onset sporadic PD. Biochemical studies support an increase in LRRK2 kinase activity and a decrease in GTPase activity for kinase domain and Roc-COR mutations, respectively. Strong evidence exists that LRRK2 toxicity is kinase dependent leading to extensive efforts to identify selective and brain-permeable LRRK2 kinase inhibitors for clinical development. Cell and animal models of PD indicate that LRRK2 mutations affect vesicular trafficking, autophagy, protein synthesis, and cytoskeletal function. Although some of these biological functions are affected consistently by most disease-linked mutations, others are not and it remains currently unclear how mutations that produce variable effects on LRRK2 biochemistry and function all commonly result in the degeneration and death of dopamine neurons. LRRK2 is typically present in Lewy bodies and its toxicity in mammalian models appears to be dependent on the presence of α-synuclein, which is elevated in human iPS-derived dopamine neurons from patients harboring LRRK2 mutations. Here, we summarize biochemical and functional studies of LRRK2 and its mutations and focus on aberrant vesicular trafficking and protein synthesis as two leading mechanisms underlying LRRK2-linked disease.
Collapse
Affiliation(s)
- Ian Martin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
143
|
Dodson MW, Leung LK, Lone M, Lizzio MA, Guo M. Novel ethyl methanesulfonate (EMS)-induced null alleles of the Drosophila homolog of LRRK2 reveal a crucial role in endolysosomal functions and autophagy in vivo. Dis Model Mech 2014; 7:1351-63. [PMID: 25288684 PMCID: PMC4257004 DOI: 10.1242/dmm.017020] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Mutations in LRRK2 cause a dominantly inherited form of Parkinson’s disease (PD) and are the most common known genetic determinant of PD. Inhibitor-based therapies targeting LRRK2 have emerged as a key therapeutic strategy in PD; thus, understanding the consequences of inhibiting the normal cellular functions of this protein is vital. Despite much interest, the physiological functions of LRRK2 remain unclear. Several recent studies have linked the toxicity caused by overexpression of pathogenic mutant forms of LRRK2 to defects in the endolysosomal and autophagy pathways, raising the question of whether endogenous LRRK2 might play a role in these processes. Here, we report the characterization of multiple novel ethyl methanesulfonate (EMS)-induced nonsense alleles in the Drosophila LRRK2 homolog, lrrk. Using these alleles, we show that lrrk loss-of-function causes striking defects in the endolysosomal and autophagy pathways, including the accumulation of markedly enlarged lysosomes that are laden with undigested contents, consistent with a defect in lysosomal degradation. lrrk loss-of-function also results in the accumulation of autophagosomes, as well as the presence of enlarged early endosomes laden with mono-ubiquitylated cargo proteins, suggesting an additional defect in lysosomal substrate delivery. Interestingly, the lysosomal abnormalities in these lrrk mutants can be suppressed by a constitutively active form of the small GTPase rab9, which promotes retromer-dependent recycling from late endosomes to the Golgi. Collectively, our data provides compelling evidence of a vital role for lrrk in lysosomal function and endolysosomal membrane transport in vivo, and suggests a link between lrrk and retromer-mediated endosomal recycling.
Collapse
Affiliation(s)
- Mark W Dodson
- Department of Neurology, University of California, Los Angeles, CA 90095, USA. Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Lok K Leung
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
| | - Mohiddin Lone
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
| | - Michael A Lizzio
- Department of Neurology, University of California, Los Angeles, CA 90095, USA. Brain Research Institute, The David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Ming Guo
- Department of Neurology, University of California, Los Angeles, CA 90095, USA. Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA. Brain Research Institute, The David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA. Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
144
|
Schapansky J, Nardozzi JD, LaVoie MJ. The complex relationships between microglia, alpha-synuclein, and LRRK2 in Parkinson's disease. Neuroscience 2014; 302:74-88. [PMID: 25284317 DOI: 10.1016/j.neuroscience.2014.09.049] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/19/2022]
Abstract
The proteins alpha-synuclein (αSyn) and leucine rich repeat kinase 2 (LRRK2) are both key players in the pathogenesis of the neurodegenerative disorder Parkinson's disease (PD), but establishing a functional link between the two proteins has proven elusive. Research studies for these two proteins have traditionally and justifiably focused in neuronal cells, but recent studies indicate that each protein could play a greater pathological role elsewhere. αSyn is expressed at high levels within neurons, but they also secrete the protein into the extracellular milieu, where it can have broad ranging effects in the nervous system and relevance to disease etiology. Similarly, low neuronal LRRK2 expression and activity suggests that LRRK2-related functions could be more relevant in cells with higher expression, such as brain-resident microglia. Microglia are monocytic immune cells that protect neurons from noxious stimuli, including pathological αSyn species, and microglial activation is believed to contribute to neuroinflammation and neuronal death in PD. Interestingly, both αSyn and LRRK2 can be linked to microglial function. Secreted αSyn can directly activate microglia, and can be taken up by microglia for clearance, while LRRK2 has been implicated in the intrinsic regulation of microglial activation and of lysosomal degradation processes. Based on these observations, the present review will focus on how PD-associated mutations in LRRK2 could potentially alter microglial biology with respect to neuronally secreted αSyn, resulting in cell dysfunction and neurodegeneration.
Collapse
Affiliation(s)
- J Schapansky
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - J D Nardozzi
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - M J LaVoie
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States.
| |
Collapse
|
145
|
Rudenko IN, Cookson MR. Heterogeneity of leucine-rich repeat kinase 2 mutations: genetics, mechanisms and therapeutic implications. Neurotherapeutics 2014; 11:738-50. [PMID: 24957201 PMCID: PMC4391379 DOI: 10.1007/s13311-014-0284-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Variation within and around the leucine-rich repeat kinase 2 (LRRK2) gene is associated with familial and sporadic Parkinson's disease (PD). Here, we discuss the prevalence of LRRK2 substitutions in different populations and their association with PD, as well as molecular and cellular mechanisms of pathologically relevant LRRK2 mutations. Kinase activation was proposed as a universal molecular mechanism for all pathogenic LRRK2 mutations, but later reports revealed heterogeneity in the effect of mutations on different activities of LRRK2. One mutation (G2019S) increases kinase activity, whereas mutations in the Ras of complex proteins (ROC)-C-terminus of ROC (COR) bidomain impair the GTPase function of LRRK2. Some risk factor variants, including G2385R in the WD40 domain, actually decrease the kinase activity of LRRK2. We suggest a model where LRRK2 mutations exert different molecular mechanisms but interfere with normal cellular function of LRRK2 at different levels of the same downstream pathway. Finally, we discuss the current state of therapeutic approaches for LRRK2-related PD.
Collapse
Affiliation(s)
- Iakov N. Rudenko
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892 USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
146
|
Novel insights into the neurobiology underlying LRRK2-linked Parkinson's disease. Neuropharmacology 2014; 85:45-56. [DOI: 10.1016/j.neuropharm.2014.05.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/16/2014] [Accepted: 05/10/2014] [Indexed: 01/08/2023]
|
147
|
p130Cas scaffolds the signalosome to direct adaptor-effector cross talk during Kaposi's sarcoma-associated herpesvirus trafficking in human microvascular dermal endothelial cells. J Virol 2014; 88:13858-78. [PMID: 25253349 DOI: 10.1128/jvi.01674-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with cell surface receptors, such as heparan sulfate, integrins (α3β1, αVβ3, and αVβ5), and EphrinA2 (EphA2), and activates focal adhesion kinase (FAK), Src, phosphoinositol 3-kinase (PI3-K), c-Cbl, and RhoA GTPase signal molecules early during lipid raft (LR)-dependent productive macropinocytic entry into human dermal microvascular endothelial cells. Our recent studies have identified CIB1 as a signal amplifier facilitating EphA2 phosphorylation and subsequent cytoskeletal cross talk during KSHV macropinocytosis. Although CIB1 lacks an enzymatic activity and traditional adaptor domain or known interacting sequence, it associated with the KSHV entry signal complex and the CIB1-KSHV association was sustained over 30 min postinfection. To identify factors scaffolding the EphA2-CIB1 signal axis, the role of major cellular scaffold protein p130Cas (Crk-associated substrate of Src) was investigated. Inhibitor and small interfering RNA (siRNA) studies demonstrated that KSHV induced p130Cas in an EphA2-, CIB1-, and Src-dependent manner. p130Cas and Crk were associated with KSHV, LRs, EphA2, and CIB1 early during infection. Live-cell microscopy and biochemical studies demonstrated that p130Cas knockdown did not affect KSHV entry but significantly reduced productive nuclear trafficking of viral DNA and routed KSHV to lysosomal degradation. p130Cas aided in scaffolding adaptor Crk to downstream guanine nucleotide exchange factor phospho-C3G possibly to coordinate GTPase signaling during KSHV trafficking. Collectively, these studies demonstrate that p130Cas acts as a bridging molecule between the KSHV-induced entry signal complex and the downstream trafficking signalosome in endothelial cells and suggest that simultaneous targeting of KSHV entry receptors with p130Cas would be an attractive potential avenue for therapeutic intervention in KSHV infection. IMPORTANCE Eukaryotic cell adaptor molecules, without any intrinsic enzymatic activity, are well known to allow a great diversity of specific and coordinated protein-protein interactions imparting signal amplification to different networks for physiological and pathological signaling. They are involved in integrating signals from growth factors, extracellular matrix molecules, bacterial pathogens, and apoptotic cells. The present study identifies human microvascular dermal endothelial (HMVEC-d) cellular scaffold protein p130Cas (Crk-associated substrate) as a platform to promote Kaposi's sarcoma-associated herpesvirus (KSHV) trafficking. Early during KSHV de novo infection, p130Cas associates with lipid rafts and scaffolds EphrinA2 (EphA2)-associated critical adaptor members to downstream effector molecules, promoting successful nuclear delivery of the KSHV genome. Hence, simultaneous targeting of the receptor EphA2 and scaffolding action of p130Cas can potentially uncouple the signal cross talk of the KSHV entry-associated upstream signal complex from the immediate downstream trafficking-associated signalosome, consequently routing KSHV toward lysosomal degradation and eventually blocking KSHV infection and associated malignancies.
Collapse
|
148
|
Cho HJ, Yu J, Xie C, Rudrabhatla P, Chen X, Wu J, Parisiadou L, Liu G, Sun L, Ma B, Ding J, Liu Z, Cai H. Leucine-rich repeat kinase 2 regulates Sec16A at ER exit sites to allow ER-Golgi export. EMBO J 2014; 33:2314-31. [PMID: 25201882 DOI: 10.15252/embj.201487807] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been associated with Parkinson's disease (PD) and other disorders. However, its normal physiological functions and pathogenic properties remain elusive. Here we show that LRRK2 regulates the anterograde ER-Golgi transport through anchoring Sec16A at the endoplasmic reticulum exit sites (ERES). LRRK2 interacted and co-localized with Sec16A, a key protein in the formation of ERES. Lrrk2 depletion caused a dispersion of Sec16A from ERES and impaired ER export. In neurons, LRRK2 and Sec16A showed extensive co-localization at the dendritic ERES (dERES) that locally regulate the transport of proteins to the dendritic spines. A loss of Lrrk2 affected the association of Sec16A with dERES and impaired the activity-dependent targeting of glutamate receptors onto the cell/synapse surface. Furthermore, the PD-related LRRK2 R1441C missense mutation in the GTPase domain interfered with the interaction of LRRK2 with Sec16A and also affected ER-Golgi transport, while LRRK2 kinase activity was not required for these functions. Therefore, our findings reveal a new physiological function of LRRK2 in ER-Golgi transport, suggesting ERES dysfunction may contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Hyun Jin Cho
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Jia Yu
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Chengsong Xie
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Parvathi Rudrabhatla
- Laboratory of Neurochemistry and Laboratory of Neurobiology National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Xi Chen
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Junbing Wu
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Loukia Parisiadou
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Guoxiang Liu
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Lixin Sun
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Bo Ma
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| | - Jinhui Ding
- Bioinformatics Core, Laboratory of Neurogenetics National Institute on Aging, Bethesda, MD, USA
| | - Zhihua Liu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases National Institutes of Health, Bethesda, MD, USA Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Huaibin Cai
- Transgenics Section, Laboratory of Neurogenetics National Institute on Aging National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
149
|
Wang X, Huang T, Bu G, Xu H. Dysregulation of protein trafficking in neurodegeneration. Mol Neurodegener 2014; 9:31. [PMID: 25152012 PMCID: PMC4237948 DOI: 10.1186/1750-1326-9-31] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/14/2014] [Indexed: 02/02/2023] Open
Abstract
Intracellular protein trafficking plays an important role in neuronal function and survival. Protein misfolding is a common theme found in many neurodegenerative diseases, and intracellular trafficking machinery contributes to the pathological accumulation and clearance of misfolded proteins. Although neurodegenerative diseases exhibit distinct pathological features, abnormal endocytic trafficking is apparent in several neurodegenerative diseases, such as Alzheimer’s disease (AD), Down syndrome (DS) and Parkinson’s disease (PD). In this review, we will focus on protein sorting defects in three major neurodegenerative diseases, including AD, DS and PD. An important pathological feature of AD is the presence of extracellular senile plaques in the brain. Senile plaques are composed of β-amyloid (Aβ) peptide aggregates. Multiple lines of evidence demonstrate that over-production/aggregation of Aβ in the brain is a primary cause of AD and attenuation of Aβ generation has become a topic of extreme interest in AD research. Aβ is generated from β-amyloid precursor protein (APP) through sequential cleavage by β-secretase and the γ-secretase complex. Alternatively, APP can be cleaved by α-secretase within the Aβ domain to release soluble APPα which precludes Aβ generation. DS patients display a strikingly similar pathology to AD patients, including the generation of neuronal amyloid plaques. Moreover, all DS patients develop an AD-like neuropathology by their 40 s. Therefore, understanding the metabolism/processing of APP and how these underlying mechanisms may be pathologically compromised is crucial for future AD and DS therapeutic strategies. Evidence accumulated thus far reveals that synaptic vesicle regulation, endocytic trafficking, and lysosome-mediated autophagy are involved in increased susceptibility to PD. Here we review current knowledge of endosomal trafficking regulation in AD, DS and PD.
Collapse
Affiliation(s)
| | | | | | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
150
|
Schapansky J, Nardozzi JD, Felizia F, LaVoie MJ. Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy. Hum Mol Genet 2014; 23:4201-14. [PMID: 24682598 PMCID: PMC4103671 DOI: 10.1093/hmg/ddu138] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/17/2014] [Accepted: 03/25/2014] [Indexed: 12/18/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial and idiopathic Parkinson's disease. However, the mechanisms for activating its physiological function are not known, hindering identification of the biological role of endogenous LRRK2. The recent discovery that LRRK2 is highly expressed in cells of the innate immune system and genetic association is a risk factor for autoimmune disorders implies an important role for LRRK2 in pathology outside of the central nervous system. Thus, an examination of endogenous LRRK2 in immune cells could provide insight into the protein's function. Here, we establish that stimulation of specific Toll-like receptors results in a complex biochemical activation of endogenous LRRK2, with early phosphorylation of LRRK2 preceding its dimerization and membrane translocation. Membrane-associated LRRK2 co-localized to autophagosome membranes following either TLR4 stimulation or mTOR inhibition with rapamycin. Silencing of endogenous LRRK2 expression resulted in deficits in the induction of autophagy and clearance of a well-described macroautophagy substrate, demonstrating the critical role of endogenous LRRK2 in regulating autophagy. Inhibition of LRRK2 kinase activity also reduced autophagic degradation and suggested the importance of the kinase domain in the regulation of autophagy. Our results demonstrate a well-orchestrated series of biochemical events involved in the activation of LRRK2 important to its physiological function. With similarities observed across multiple cell types and stimuli, these findings are likely relevant in all cell types that natively express endogenous LRRK2, and provide insights into LRRK2 function and its role in human disease.
Collapse
Affiliation(s)
- Jason Schapansky
- Center for Neurologic Diseases, Harvard Medical School, Boston, MA 02115, USA and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jonathan D Nardozzi
- Center for Neurologic Diseases, Harvard Medical School, Boston, MA 02115, USA and Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Matthew J LaVoie
- Center for Neurologic Diseases, Harvard Medical School, Boston, MA 02115, USA and Brigham and Women's Hospital, Boston, MA 02115, USA
| |
Collapse
|