1
|
Hyderi Z, Farhana M S, Singh TP, Ravi AV. Therapeutic Targeting of Autosomal Parkinson's Disease by Modulation of Leucine-Rich Repeat Kinase 2 (LRRK2) Protein. Brain Res 2025; 1860:149674. [PMID: 40345365 DOI: 10.1016/j.brainres.2025.149674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/18/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Leucine-Rich Repeat Kinase 2 (LRRK2) is gaining attention as a key therapeutic target for autosomal dominant Parkinson's disease (PD). The primary genetic aetiology of familial PD, accounting for around 5-6 % of familial cases and 2 % of sporadic cases, is mutations in the LRRK2 gene. The most prevalent mutation, G2019S, increases kinase activity, which phosphorylates important serine residues that control LRRK2 function, such as Ser910 and Ser935, leading to the development of PD. The development of LRRK2 inhibitors has emerged as a key area of study for PD therapy. In preclinical research, these inhibitors have demonstrated promise in reducing PD-related damage by altering the cellular localisation of LRRK2 and reduced phosphorylation. In addition to kinase action, LRRK2 is involved in autophagy and mitochondrial function. This participation implies that PD markers including mitochondrial dysfunction and defective autophagy may be addressed by LRRK2-targeted treatments. Moreover, selective LRRK2 inhibitors show promise in the treatment of PD, and more research into the molecular role of LRRK2 in PD is essential to developing efficient therapies that will improve patient outcomes and reduce the course of the illness. This review discusses the role of LRRK2 in pathogenesis of PD and current treatment approaches, particularly LRRK2 kinase inhibitors, and their potential to slow disease progression, along with recent advancements in clinical trials and future outlooks for improving outcomes in PD.
Collapse
Affiliation(s)
- Zeeshan Hyderi
- Lab in Microbiology and Marine Biotechnology, Department of Biotechnology, Alagappa University, Karaikudi 630003, India
| | - Shirin Farhana M
- Department of Biotechnology, Alagappa University, Karaikudi 630003, India
| | - Tej Pal Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Arumugam Veera Ravi
- Lab in Microbiology and Marine Biotechnology, Department of Biotechnology, Alagappa University, Karaikudi 630003, India.
| |
Collapse
|
2
|
Kulasooriya S, Liu H, Vijayakumar S, Bloom C, Tu S, Borgmeier BJ, Zhou M, Tao L, Kachar B, He DZ. Single-cell Transcriptomics Unravel Stereocilia Degeneration as a Key Contributor to Age-related Vestibular Dysfunction in Mice and Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636113. [PMID: 39975332 PMCID: PMC11838431 DOI: 10.1101/2025.02.02.636113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Age-related vestibular dysfunction (ARVD) is a prevalent, debilitating condition in the elderly. The etiology and molecular mechanisms are poorly understood. We focused on mechanosensitive hair cells (HCs) as they are particularly vulnerable to aging. Using single-cell RNA-seq transcriptomes of young and old mouse vestibular HCs, we show that aging HCs display both universal molecular blueprints, such as genomic instability, mitochondrial dysfunction, and impaired proteostasis, and cell type-specific aging signatures associated with deterioration of hair bundles and mechanotransduction. These signatures are also observed in aged human vestibular HCs, suggesting shared mechanisms. Importantly, morphological and functional analysis revealed that bundle degeneration and vestibular functional decline precede HC loss, highlighting the deterioration of mechanotransduction as a key contributor to ARVD. Furthermore, molecular and cellular changes associated with aging signatures are less pronounced in vestibular HCs than in cochlear HCs, underscoring the different pace of aging between the two mammalian inner ear sensory epithelia.
Collapse
|
3
|
Ashraf D, Khan MR, Dawson TM, Dawson VL. Protein Translation in the Pathogenesis of Parkinson's Disease. Int J Mol Sci 2024; 25:2393. [PMID: 38397070 PMCID: PMC10888601 DOI: 10.3390/ijms25042393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, research into Parkinson's disease and similar neurodegenerative disorders has increasingly suggested that these conditions are synonymous with failures in proteostasis. However, the spotlight of this research has remained firmly focused on the tail end of proteostasis, primarily aggregation, misfolding, and degradation, with protein translation being comparatively overlooked. Now, there is an increasing body of evidence supporting a potential role for translation in the pathogenesis of PD, and its dysregulation is already established in other similar neurodegenerative conditions. In this paper, we consider how altered protein translation fits into the broader picture of PD pathogenesis, working hand in hand to compound the stress placed on neurons, until this becomes irrecoverable. We will also consider molecular players of interest, recent evidence that suggests that aggregates may directly influence translation in PD progression, and the implications for the role of protein translation in our development of clinically useful diagnostics and therapeutics.
Collapse
Affiliation(s)
- Daniyal Ashraf
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.A.); (M.R.K.)
- School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 111, Cambridge CB2 0SP, UK
| | - Mohammed Repon Khan
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.A.); (M.R.K.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.A.); (M.R.K.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
- Solomon H. Snyder Department of Neuroscience, 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
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.A.); (M.R.K.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Patel A, Patel S, Mehta M, Patel Y, Langaliya D, Bhalodiya S, Bambharoliya T. Recent Update on the Development of Leucine- Rich Repeat Kinase 2 (LRRK2) Inhibitors: A Promising Target for the Treatment of Parkinson's Disease. Med Chem 2022; 18:757-771. [PMID: 35168510 DOI: 10.2174/1573406418666220215122136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease is a relatively common neurological disorder with incidence increasing with age. Since current medications only relieve the symptoms and do not change the course of the disease, therefore, finding disease-modifying therapies is a critical unmet medical need. However, significant progress in understanding how genetics underpins Parkinson's disease (PD) has opened up new opportunities for understanding disease pathogenesis and identifying possible therapeutic targets. One such target is leucine-rich repeat kinase 2 (LRRK2), an elusive enzyme implicated in both familial and idiopathic PD risk. As a result, both academia and industry have promoted the development of potent and selective inhibitors of LRRK2. In this review, we have summarized recent progress on the discovery and development of LRKK2 inhibitors as well as the bioactivity of several small-molecule LRRK2 inhibitors that have been used to inhibit LRRK2 kinase activity in vitro or in vivo.
Collapse
Affiliation(s)
- Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Stuti Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Meshwa Mehta
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Yug Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Dhruv Langaliya
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | - Shyam Bhalodiya
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT-Campus, Changa-388421, Anand, Gujarat, India
| | | |
Collapse
|
5
|
Pischedda F, Cirnaru MD, Ponzoni L, Sandre M, Biosa A, Carrion MP, Marin O, Morari M, Pan L, Greggio E, Bandopadhyay R, Sala M, Piccoli G. LRRK2 G2019S kinase activity triggers neurotoxic NSF aggregation. Brain 2021; 144:1509-1525. [PMID: 33876242 DOI: 10.1093/brain/awab073] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/11/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease is characterized by the progressive degeneration of dopaminergic neurons within the substantia nigra pars compacta and the presence of protein aggregates in surviving neurons. The LRRK2 G2019S mutation is one of the major determinants of familial Parkinson's disease cases and leads to late-onset Parkinson's disease with pleomorphic pathology, including α-synuclein accumulation and deposition of protein inclusions. We demonstrated that LRRK2 phosphorylates N-ethylmaleimide sensitive factor (NSF). We observed aggregates containing NSF in basal ganglia specimens from patients with Parkinson's disease carrying the G2019S variant, and in cellular and animal models expressing the LRRK2 G2019S variant. We found that LRRK2 G2019S kinase activity induces the accumulation of NSF in toxic aggregates. Of note, the induction of autophagy cleared NSF aggregation and rescued motor and cognitive impairment observed in aged hG2019S bacterial artificial chromosome (BAC) mice. We suggest that LRRK2 G2019S pathological phosphorylation impacts on NSF biochemical properties, thus causing the formation of cytotoxic protein inclusions.
Collapse
Affiliation(s)
- Francesca Pischedda
- CIBIO, Università degli Studi di Trento, Trento, Italy.,Dulbecco Telethon Institute, Rome, Italy
| | | | | | - Michele Sandre
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Alice Biosa
- Department of Biology, University of Padova, Padova, Italy
| | - Maria Perez Carrion
- CIBIO, Università degli Studi di Trento, Trento, Italy.,Unidad Asociada Neurodeath, Faculty of Medicine, University of Castilla-La Mancha, 02008, Albacete, Spain
| | - Oriano Marin
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Michele Morari
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Lifeng Pan
- Shanghai Institute of Organic Chemistry, Shanghai, China
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies and Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | | | - Giovanni Piccoli
- CIBIO, Università degli Studi di Trento, Trento, Italy.,Dulbecco Telethon Institute, Rome, Italy
| |
Collapse
|
6
|
Pathogenic LRRK2 requires secondary factors to induce cellular toxicity. Biosci Rep 2021; 40:226517. [PMID: 32975566 PMCID: PMC7560525 DOI: 10.1042/bsr20202225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 12/02/2022] Open
Abstract
Pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene belong to the most common genetic causes of inherited Parkinson’s disease (PD) and variations in its locus increase the risk to develop sporadic PD. Extensive research efforts aimed at understanding how changes in the LRRK2 function result in molecular alterations that ultimately lead to PD. Cellular LRRK2-based models revealed several potential pathophysiological mechanisms including apoptotic cell death, LRRK2 protein accumulation and deficits in neurite outgrowth. However, highly variable outcomes between different cellular models have been reported. Here, we have investigated the effect of different experimental conditions, such as the use of different tags and gene transfer methods, in various cellular LRRK2 models. Readouts included cell death, sensitivity to oxidative stress, LRRK2 relocalization, α-synuclein aggregation and neurite outgrowth in cell culture, as well as neurite maintenance in vivo. We show that overexpression levels and/or the tag fused to LRRK2 affect the relocalization of LRRK2 to filamentous and skein-like structures. We found that overexpression of LRRK2 per se is not sufficient to induce cellular toxicity or to affect α-synuclein-induced toxicity and aggregate formation. Finally, neurite outgrowth/retraction experiments in cell lines and in vivo revealed that secondary, yet unknown, factors are required for the pathogenic LRRK2 effects on neurite length. Our findings stress the importance of technical and biological factors in LRRK2-induced cellular phenotypes and hence imply that conclusions based on these types of LRRK2-based assays should be interpreted with caution.
Collapse
|
7
|
Fang CY, Lai TC, Hsiao M, Chang YC. The Diverse Roles of TAO Kinases in Health and Diseases. Int J Mol Sci 2020; 21:E7463. [PMID: 33050415 PMCID: PMC7589832 DOI: 10.3390/ijms21207463] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Thousand and one kinases (TAOKs) are members of the MAP kinase kinase kinase (MAP3K) family. Three members of this subfamily, TAOK1, 2, and 3, have been identified in mammals. It has been shown that TAOK1, 2 and 3 regulate the p38 MAPK and Hippo signaling pathways, while TAOK 1 and 2 modulate the SAPK/JNK cascade. Furthermore, TAOKs are involved in additional interactions with other cellular proteins and all of these pathways modulate vital physiological and pathophysiological responses in cells and tissues. Dysregulation of TAOK-related pathways is implicated in the development of diseases including inflammatory and immune disorders, cancer and drug resistance, and autism and Alzheimer's diseases. This review collates current knowledge concerning the roles of TAOKs in protein-protein interaction, signal transduction, physiological regulation, and pathogenesis and summarizes the recent development of TAOK-specific inhibitors that have the potential to ameliorate TAOKs' effects in pathological situations.
Collapse
Affiliation(s)
- Chih-Yeu Fang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Tsung-Ching Lai
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang-Ming University, Taipei 112, Taiwan
| |
Collapse
|
8
|
LRRK2, alpha-synuclein, and tau: partners in crime or unfortunate bystanders? Biochem Soc Trans 2019; 47:827-838. [PMID: 31085616 DOI: 10.1042/bst20180466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
The identification of genetic forms of Parkinson's disease (PD) has tremendously expanded our understanding of the players and mechanisms involved. Mutations in the genes encoding for alpha-synuclein (aSyn), LRRK2, and tau have been associated with familial and sporadic forms of the disease. aSyn is the major component of Lewy bodies and Lewy neurites, which are pathognomonic protein inclusions in PD. Hyperphosphorylated tau protein accumulates in neurofibrillary tangles in the brains of Alzheimer's disease patients but is also seen in the brains of PD patients. LRRK2 is a complex multi-domain protein with kinase and GTPase enzymatic activity. Since aSyn and tau are phosphoproteins, we review the possible interplay between the three proteins. Understanding the interplay between LRRK2, aSyn and tau is extremely important, as this may enable the identification of novel targets and pathways for therapeutic intervention.
Collapse
|
9
|
Pajarillo E, Rizor A, Lee J, Aschner M, Lee E. The role of posttranslational modifications of α-synuclein and LRRK2 in Parkinson's disease: Potential contributions of environmental factors. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1992-2000. [PMID: 30481588 PMCID: PMC6534484 DOI: 10.1016/j.bbadis.2018.11.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD), and the most prevalent movement disorder. PD is characterized by dopaminergic neurodegeneration in the substantia nigra, but its etiology has yet to be established. Among several genetic variants contributing to PD pathogenesis, α-synuclein and leucine-rich repeat kinase (LRRK2) are widely associated with neuropathological phenotypes in familial and sporadic PD. α-Synuclein and LRRK2 found in Lewy bodies, a pathogenic hallmark of PD, are often posttranslationally modified. As posttranslational modifications (PTMs) are key processes in regulating the stability, localization, and function of proteins, PTMs have emerged as important modulators of α-synuclein and LRRK2 pathology. Aberrant PTMs altering phosphorylation, ubiquitination, nitration and truncation of these proteins promote PD pathogenesis, while other PTMs such as sumoylation may be protective. Although the causes of many aberrant PTMs are unknown, environmental risk factors may contribute to their aberrancy. Environmental toxicants such as rotenone and paraquat have been shown to interact with these proteins and promote their abnormal PTMs. Notably, manganese (Mn) exposure leads to a PD-like neurological disorder referred to as manganism-and induces pathogenic PTMs of α-synuclein and LRRK2. In this review, we highlight the role of PTMs of α-synuclein and LRRK2 in PD pathogenesis and discuss the impact of environmental risk factors on their aberrancy.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, United States of America
| | - Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, United States of America
| | - Jayden Lee
- Department of Speech, Language & Hearing Sciences, Boston University, Boston, MA 02215, United States of America
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States of America
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, United States of America.
| |
Collapse
|
10
|
Cryo-EM analysis of homodimeric full-length LRRK2 and LRRK1 protein complexes. Sci Rep 2017; 7:8667. [PMID: 28819229 PMCID: PMC5561129 DOI: 10.1038/s41598-017-09126-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/03/2017] [Indexed: 11/30/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain protein implicated in the pathogenesis of both familial and sporadic Parkinson’s disease (PD), and currently one of the most promising therapeutic targets for drug design in Parkinson’s disease. In contrast, LRRK1, the closest homologue to LRRK2, does not play any role in PD. Here, we use cryo-electron microscopy (cryo-EM) and single particle analysis to gain structural insight into the full-length dimeric structures of LRRK2 and LRRK1. Differential scanning fluorimetry-based screening of purification buffers showed that elution of the purified LRRK2 protein in a high pH buffer is beneficial in obtaining high quality cryo-EM images. Next, analysis of the 3D maps generated from the cryo-EM data show 16 and 25 Å resolution structures of full length LRRK2 and LRRK1, respectively, revealing the overall shape of the dimers with two-fold symmetric orientations of the protomers that is closely similar between the two proteins. These results suggest that dimerization mechanisms of both LRRKs are closely related and hence that specificities in functions of each LRRK are likely derived from LRRK2 and LRRK1’s other biochemical functions. To our knowledge, this study is the first to provide 3D structural insights in LRRK2 and LRRK1 dimers in parallel.
Collapse
|
11
|
Current Opinion on the Role of Neurogenesis in the Therapeutic Strategies for Alzheimer Disease, Parkinson Disease, and Ischemic Stroke; Considering Neuronal Voiding Function. Int Neurourol J 2016; 20:276-287. [PMID: 28043116 PMCID: PMC5209581 DOI: 10.5213/inj.1632776.388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/12/2016] [Indexed: 01/01/2023] Open
Abstract
Neurological diseases such as Alzheimer, Parkinson, and ischemic stroke have increased in occurrence and become important health issues throughout the world. There is currently no effective therapeutic strategy for addressing neurological deficits after the development of these major neurological disorders. In recent years, it has become accepted that adult neural stem cells located in the subventricular and subgranular zones have the ability to proliferate and differentiate in order to replace lost or damaged neural cells. There have been many limitations in the clinical application of both endogenous and exogenous neurogenesis for neurological disorders. However, many studies have investigated novel mechanisms in neurogenesis and have shown that these limitations can potentially be overcome with appropriate stimulation and various approaches. We will review concepts related to possible therapeutic strategies focused on the perspective of neurogenesis for the treatment of patients diagnosed with Alzheimer disease, Parkinson disease, and ischemic stroke based on current reports.
Collapse
|
12
|
Schwab AJ, Ebert AD. Neurite Aggregation and Calcium Dysfunction in iPSC-Derived Sensory Neurons with Parkinson's Disease-Related LRRK2 G2019S Mutation. Stem Cell Reports 2016; 5:1039-1052. [PMID: 26651604 PMCID: PMC4682343 DOI: 10.1016/j.stemcr.2015.11.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/11/2015] [Accepted: 11/12/2015] [Indexed: 01/15/2023] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most-common genetic determinants of Parkinson’s disease (PD). The G2019S mutation is detected most frequently and is associated with increased kinase activity. Whereas G2019S mutant dopamine neurons exhibit neurite elongation deficits, the effect of G2019S on other neuronal subtypes is unknown. As PD patients also suffer from non-motor symptoms that may be unrelated to dopamine neuron loss, we used induced pluripotent stem cells (iPSCs) to assess morphological and functional properties of peripheral sensory neurons. LRRK2 G2019S iPSC-derived sensory neurons exhibited normal neurite length but had large microtubule-containing neurite aggregations. Additionally, LRRK2 G2019S iPSC-derived sensory neurons displayed altered calcium dynamics. Treatment with LRRK2 kinase inhibitors resulted in significant, but not complete, morphological and functional rescue. These data indicate a role for LRRK2 kinase activity in sensory neuron structure and function, which when disrupted, may lead to sensory neuron deficits in PD. LRRK2 iPSC sensory neurons show neurite aggregations and abnormal calcium dynamics LRRK2 iPSC sensory neuron defects are distinct from the dopamine neuron defects Kinase inhibition of LRRK2 partially restored sensory neuron structure and function Abnormal sensory neuron phenotypes may relate to non-motor symptoms observed in PD
Collapse
Affiliation(s)
- Andrew J Schwab
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
13
|
Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci 2016; 74:409-434. [PMID: 27600680 PMCID: PMC5241350 DOI: 10.1007/s00018-016-2351-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
Cytoskeletal homeostasis is essential for the development, survival and maintenance of an efficient nervous system. Microtubules are highly dynamic polymers important for neuronal growth, morphology, migration and polarity. In cooperation with several classes of binding proteins, microtubules regulate long-distance intracellular cargo trafficking along axons and dendrites. The importance of a delicate interplay between cytoskeletal components is reflected in several human neurodegenerative disorders linked to abnormal microtubule dynamics, including Parkinson’s disease (PD). Mounting evidence now suggests PD pathogenesis might be underlined by early cytoskeletal dysfunction. Advances in genetics have identified PD-associated mutations and variants in genes encoding various proteins affecting microtubule function including the microtubule-associated protein tau. In this review, we highlight the role of microtubules, their major posttranslational modifications and microtubule associated proteins in neuronal function. We then present key evidence on the contribution of microtubule dysfunction to PD. Finally, we discuss how regulation of microtubule dynamics with microtubule-targeting agents and deacetylase inhibitors represents a promising strategy for innovative therapeutic development.
Collapse
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Yu WJ, Cheng L, Li NN, Wang L, Tan EK, Peng R. Interaction between SNCA, LRRK2 and GAK increases susceptibility to Parkinson's disease in a Chinese population. eNeurologicalSci 2015; 1:3-6. [PMID: 29479569 PMCID: PMC5852682 DOI: 10.1016/j.ensci.2015.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 02/05/2023] Open
Abstract
PD is a complex disease, and may result from gene–gene and gene–environment interactions. There are limited studies on gene–gene interactions in PD. We and others have previously shown that SNCA rs356219, LRRK2 (rs2046932 and rs7304279) and GAK (rs1564282) are risk factors in sporadic PD. Since the expression of SNCA and neurotoxicity of alpha-synuclein are affected by LRRK2 and GAK, we hypothesize that their genetic risk variants may interact with each other. Here we investigated the interaction of SNCA rs356219, LRRK2rs7304279 and rs2046932 and GAK rs1564282 using the Multifactor Dimensionality Reduction (MDR) in a Chinese PD patient–control series (534 patients and 435 controls) and the cumulative risk effect of SNCA, LRRK2 and GAK. The MDR analysis showed a significant gene–gene interaction between the rs356219 of SNCA, rs2046932 of LRRK2 and rs1564282 of GAK. Moreover, individuals with increasing numbers of variants had an increasing likelihood of having PD, compared with those carrying none of the variants. The estimated OR for developing PD in individuals carrying 3 variants was 5.89. We demonstrated for the first time that SNPs in SNCA, LRRK2 and GAK interacted with each other to confer an increased risk of PD. In addition, PD risk increased cumulatively with the increasing number of variants.
Collapse
Affiliation(s)
- Wen-Juan Yu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Lan Cheng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Nan-Nan Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Ling Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Eng-King Tan
- Duke-NUS Graduate Medical School, Singapore, Singapore.,Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Singapore, Singapore
| | - Rong Peng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
18
|
Deregulation of protein translation control, a potential game-changing hypothesis for Parkinson's disease pathogenesis. Trends Mol Med 2015; 21:466-72. [PMID: 26091824 DOI: 10.1016/j.molmed.2015.05.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/15/2015] [Accepted: 05/19/2015] [Indexed: 02/08/2023]
Abstract
Protein translation is one of the most fundamental and exquisitely controlled processes in biology, and is energetically demanding. The deregulation of this process is deleterious to cells, as demonstrated by several diseases caused by mutations in protein translation machinery. Emerging evidence now points to a role for protein translation in the pathogenesis of Parkinson's disease (PD); a debilitating neurodegenerative movement disorder. In this paper, we propose a hypothesis that protein translation machinery, PD-associated proteins and PD pathology are connected in a functional network linking cell survival to protein translation control. This hypothesis is a potential game changer in the field of the molecular pathogenesis of PD, with implications for the development of PD diagnostics and disease-modifying therapies.
Collapse
|
19
|
LRRK2 Promotes Tau Accumulation, Aggregation and Release. Mol Neurobiol 2015; 53:3124-3135. [PMID: 26014385 DOI: 10.1007/s12035-015-9209-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/04/2015] [Indexed: 01/09/2023]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are known as the most frequent cause of familial Parkinson's disease (PD), but are also present in sporadic cases. The G2019S-LRRK2 mutation is located in the kinase domain of the protein, and has consistently been reported to promote a gain of kinase function. Several proteins have been reported as LRRK2 substrates and/or interactors, suggesting possible pathways involved in neurodegeneration in PD. Hyperphosphorylated Tau protein accumulates in neurofibrillary tangles, a typical pathological hallmark in Alzheimer's disease and frontotemporal dementia. In addition, it is also frequently found in the brains of PD patients. Although LRRK2 is a kinase, it appears that a putative interaction with Tau is phosphorylation-independent. However, the underlying mechanisms and the cellular consequences of this interaction are still unclear. In this study, we demonstrate an interaction between LRRK2 and Tau and that LRRK2 promotes the accumulation of non-monomeric and high-molecular weight (HMW) Tau species independent of its kinase activity. Interestingly, we found that LRRK2 increases Tau secretion, possibly as a consequence of an impairment of Tau proteasomal degradation. Our data highlight a mechanism through which LRRK2 regulates intracellular Tau levels, contributing to the progression of the pathology caused by the LRRK2-mediated proteasome impairment. In total, our findings suggest that the interplay between LRRK2 and proteasome activity might constitute a valid target for therapeutic intervention in PD.
Collapse
|
20
|
Krumova P, Reyniers L, Meyer M, Lobbestael E, Stauffer D, Gerrits B, Muller L, Hoving S, Kaupmann K, Voshol J, Fabbro D, Bauer A, Rovelli G, Taymans JM, Bouwmeester T, Baekelandt V. Chemical genetic approach identifies microtubule affinity-regulating kinase 1 as a leucine-rich repeat kinase 2 substrate. FASEB J 2015; 29:2980-92. [PMID: 25854701 DOI: 10.1096/fj.14-262329] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/09/2015] [Indexed: 12/25/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of autosomal-dominant forms of Parkinson's disease. LRRK2 is a modular, multidomain protein containing 2 enzymatic domains, including a kinase domain, as well as several protein-protein interaction domains, pointing to a role in cellular signaling. Although enormous efforts have been made, the exact pathophysiologic mechanisms of LRRK2 are still not completely known. In this study, we used a chemical genetics approach to identify LRRK2 substrates from mouse brain. This approach allows the identification of substrates of 1 particular kinase in a complex cellular environment. Several of the identified peptides are involved in the regulation of microtubule (MT) dynamics, including microtubule-associating protein (MAP)/microtubule affinity-regulating kinase 1 (MARK1). MARK1 is a serine/threonine kinase known to phosphorylate MT-binding proteins such as Tau, MAP2, and MAP4 at KXGS motifs leading to MT destabilization. In vitro kinase assays and metabolic-labeling experiments in living cells confirmed MARK1 as an LRRK2 substrate. Moreover, we also showed that LRRK2 and MARK1 are interacting in eukaryotic cells. Our findings contribute to the identification of physiologic LRRK2 substrates and point to a potential mechanism explaining the reported effects of LRRK2 on neurite morphology.
Collapse
Affiliation(s)
- Petranka Krumova
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Lauran Reyniers
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Marc Meyer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Evy Lobbestael
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Daniela Stauffer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Bertran Gerrits
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Lionel Muller
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sjouke Hoving
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Klemens Kaupmann
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Johannes Voshol
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Doriano Fabbro
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Andreas Bauer
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Giorgio Rovelli
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Jean-Marc Taymans
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Tewis Bouwmeester
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- *Novartis Institutes for Biomedical Research, Basel, Switzerland; and Department of Neurosciences, Research Group for Neurobiology and Gene Therapy, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Taymans JM, Baekelandt V. Phosphatases of α-synuclein, LRRK2, and tau: important players in the phosphorylation-dependent pathology of Parkinsonism. Front Genet 2014; 5:382. [PMID: 25426138 PMCID: PMC4224088 DOI: 10.3389/fgene.2014.00382] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
An important challenge in the field of Parkinson’s disease (PD) is to develop disease modifying therapies capable of stalling or even halting disease progression. Coupled to this challenge is the need to identify disease biomarkers, in order to identify pre-symptomatic hallmarks of disease and monitor disease progression. The answer to these challenges lies in the elucidation of the molecular causes underlying PD, for which important leads are disease genes identified in studies investigating the underlying genetic causes of PD. LRRK2 and α-syn have been both linked to familial forms of PD as well as associated to sporadic PD. Another gene, microtubule associated protein tau (MAPT), has been genetically linked to a dominant form of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and genome-wide association studies report a strong association between MAPT and sporadic PD. Interestingly, LRRK2, α-syn, and tau are all phosphorylated proteins, and their phosphorylation patterns are linked to disease. In this review, we provide an overview of the evidence linking LRRK2, α-syn, and tau phosphorylation to PD pathology and focus on studies which have identified phosphatases responsible for dephosphorylation of pathology-related phosphorylations. We also discuss how the LRRK2, α-syn, and tau phosphatases may point to separate or cross-talking pathological pathways in PD. Finally, we will discuss how the study of phosphatases of dominant Parkinsonism proteins opens perspectives for targeting pathological phosphorylation events.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| |
Collapse
|
22
|
Johnson WM, Yao C, Siedlak SL, Wang W, Zhu X, Caldwell GA, Wilson-Delfosse AL, Mieyal JJ, Chen SG. Glutaredoxin deficiency exacerbates neurodegeneration in C. elegans models of Parkinson's disease. Hum Mol Genet 2014; 24:1322-35. [PMID: 25355420 DOI: 10.1093/hmg/ddu542] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is characterized by selective degeneration of dopaminergic neurons. Although the etiology of PD remains incompletely understood, oxidative stress has been implicated as an important contributor in the development of PD. Oxidative stress can lead to oxidation and functional perturbation of proteins critical to neuronal survival. Glutaredoxin 1 (Grx1) is an evolutionally conserved antioxidant enzyme that repairs protein oxidation by reversing the oxidative modification of cysteine known as S-glutathionylation. We aimed to explore the regulatory role of Grx1 in PD. We first examined the levels of Grx1 in postmortem midbrain samples from PD patients, and observed that Grx1 content is decreased in PD, specifically within the dopaminergic neurons. We subsequently investigated the potential role of Grx1 deficiency in PD pathogenesis by examining the consequences of loss of the Caenorhabditis elegans Grx1 homolog in well-established worm models of familial PD caused by overexpression of pathogenic human LRRK2 mutants G2019S or R1441C. We found that loss of the Grx1 homolog led to significant exacerbation of the neurodegenerative phenotype in C. elegans overexpressing the human LRRK2 mutants. Re-expression in the dopaminergic neurons of the active, but not a catalytically inactive form of the Grx1 homolog rescued the exacerbated phenotype. Loss of the Grx1 homolog also exacerbated the neurodegenerative phenotype in other C. elegans models, including overexpression of human α-synuclein and overexpression of tyrosine hydroxylase (a model of sporadic PD). Therefore, our results reveal a novel neuroprotective role of glutaredoxin against dopaminergic neurodegeneration in models of familial and sporadic PD.
Collapse
Affiliation(s)
| | - Chen Yao
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wenzhang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | | | - John J Mieyal
- Department of Pharmacology and Louis B. Stokes Veterans Affairs Medical Research Center, Cleveland, OH 44106, USA
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA,
| |
Collapse
|
23
|
Indolinone based LRRK2 kinase inhibitors with a key hydrogen bond. Bioorg Med Chem Lett 2014; 24:4630-4637. [PMID: 25219901 DOI: 10.1016/j.bmcl.2014.08.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 11/22/2022]
Abstract
The most prevalent leucine-rich repeat kinase 2 (LRRK2) mutation G2019S is associated with Parkinson's disease (PD). It enhances kinase activity and has been identified in both familial and sporadic cases. Kinase activity was reported to be required for LRRK2 mutants to exert their toxic effects. Hence LRRK2 kinase inhibition may be a promising therapeutic target for PD. Here we report on the discovery and characterization of indolinone based LRRK2 inhibitors. Indolinone 15b, the most potent and selective inhibitor of the present series, is characterized by an IC50 of 15nM against wild-type LRRK2 and 10nM against the LRRK2 G2019S mutant, respectively. Compound 15b was further evaluated in a kinase panel including 46 human protein kinases and in a zebrafish embryo phenotype assay, which enabled toxicity determination in whole organisms.
Collapse
|
24
|
Genetic and pharmacological evidence that G2019S LRRK2 confers a hyperkinetic phenotype, resistant to motor decline associated with aging. Neurobiol Dis 2014; 71:62-73. [PMID: 25107341 PMCID: PMC4194318 DOI: 10.1016/j.nbd.2014.07.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 07/09/2014] [Accepted: 07/28/2014] [Indexed: 11/22/2022] Open
Abstract
The leucine-rich repeat kinase 2 mutation G2019S in the kinase-domain is the most common genetic cause of Parkinson's disease. To investigate the impact of the G2019S mutation on motor activity in vivo, a longitudinal phenotyping approach was developed in knock-in (KI) mice bearing this kinase-enhancing mutation. Two cohorts of G2019S KI mice and wild-type littermates (WT) were subjected to behavioral tests, specific for akinesia, bradykinesia and overall gait ability, at different ages (3, 6, 10, 15 and 19 months). The motor performance of G2019S KI mice remained stable up to the age of 19 months and did not show the typical age-related decline in immobility time and stepping activity of WT. Several lines of evidence suggest that enhanced LRRK2 kinase activity is the main contributor to the observed hyperkinetic phenotype of G2019S KI mice: i) KI mice carrying a LRRK2 kinase-dead mutation (D1994S KD) showed a similar progressive motor decline as WT; ii) two LRRK2 kinase inhibitors, H-1152 and Nov-LRRK2-11, acutely reversed the hyperkinetic phenotype of G2019S KI mice, while being ineffective in WT or D1994S KD animals. LRRK2 target engagement in vivo was further substantiated by reduction of LRRK2 phosphorylation at Ser935 in the striatum and cortex at efficacious doses of Nov-LRRK2-11, and in the striatum at efficacious doses of H-1152. In summary, expression of the G2019S mutation in the mouse LRRK2 gene confers a hyperkinetic phenotype that is resistant to age-related motor decline, likely via enhancement of LRRK2 kinase activity. This study provides an in vivo model to investigate the effects of LRRK2 inhibitors on motor function. The LRRK2 G2019S mutation confers a hyperkinetic phenotype. The LRRK2 D1994S kinase-dead mutation does not affect motor phenotype. The LRRK2 kinase inhibitors reverse motor phenotype of G2019S mice. The LRRK2 kinase inhibitors inhibit LRRK2 phosphorylation at Ser935 ex-vivo.
Collapse
|
25
|
Plowey ED, Johnson JW, Steer E, Zhu W, Eisenberg DA, Valentino NM, Liu YJ, Chu CT. Mutant LRRK2 enhances glutamatergic synapse activity and evokes excitotoxic dendrite degeneration. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1596-603. [PMID: 24874075 DOI: 10.1016/j.bbadis.2014.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/12/2014] [Accepted: 05/19/2014] [Indexed: 01/01/2023]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2), which are associated with autosomal dominant Parkinson's disease, elicit progressive dendrite degeneration in neurons. We hypothesized that synaptic dysregulation contributes to mutant LRRK2-induced dendritic injury. We performed in vitro whole-cell voltage clamp studies of glutamatergic receptor agonist responses and glutamatergic synaptic activity in cultured rat cortical neurons expressing full-length wild-type and mutant forms of LRRK2. Expression of the pathogenic G2019S or R1441C LRRK2 mutants resulted in larger whole-cell current responses to direct application of AMPA and NMDA receptor agonists. In addition, mutant LRRK2-expressing neurons exhibited an increased frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs) in conjunction with increased excitatory synapse density as assessed by immunofluorescence for PSD95 and VGLUT1. Mutant LRRK2-expressing neurons showed enhanced vulnerability to acute synaptic glutamate stress. Furthermore, treatment with the NMDA receptor antagonist memantine significantly protected against subsequent losses in dendrite length and branching complexity. These data demonstrate an early association between mutant LRRK2 and increased excitatory synapse activity, implicating an excitotoxic contribution to mutant LRRK2 induced dendrite degeneration.
Collapse
Affiliation(s)
- Edward D Plowey
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jon W Johnson
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erin Steer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wan Zhu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - David A Eisenberg
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Yong-Jian Liu
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Charleen T Chu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Pchelina SN, Emelyanov AK, Usenko TS. Molecular basis of Parkinsons’s disease linked to LRRK2 mutations. Mol Biol 2014. [DOI: 10.1134/s0026893314010117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
27
|
Abstract
Wingless/Int (Wnt) signaling pathways are signal transduction mechanisms that have been widely studied in the field of embryogenesis. Recent work has established a critical role for these pathways in brain development, especially of midbrain dopaminergic neurones. However, the fundamental importance of Wnt signaling for the normal function of mature neurones in the adult central nervous system has also lately been demonstrated by an increasing number of studies. Parkinson's disease (PD) is the second most prevalent neurodegenerative disease worldwide and is currently incurable. This debilitating disease is characterized by the progressive loss of a subset of midbrain dopaminergic neurones in the substantia nigra leading to typical extrapyramidal motor symptoms. The aetiology of PD is poorly understood but work performed over the last two decades has identified a growing number of genetic defects that underlie this condition. Here we review a growing body of data connecting genes implicated in PD--most notably the PARK genes--with Wnt signaling. These observations provide clues to the normal function of these proteins in healthy neurones and suggest that deregulated Wnt signaling might be a frequent pathomechanism leading to PD. These observations have implications for the pathogenesis and treatment of neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Daniel C. Berwick
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| |
Collapse
|
28
|
Taymans JM, Gao F, Baekelandt V. Metabolic labeling of leucine rich repeat kinases 1 and 2 with radioactive phosphate. J Vis Exp 2013:e50523. [PMID: 24084685 DOI: 10.3791/50523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Leucine rich repeat kinases 1 and 2 (LRRK1 and LRRK2) are paralogs which share a similar domain organization, including a serine-threonine kinase domain, a Ras of complex proteins domain (ROC), a C-terminal of ROC domain (COR), and leucine-rich and ankyrin-like repeats at the N-terminus. The precise cellular roles of LRRK1 and LRRK2 have yet to be elucidated, however LRRK1 has been implicated in tyrosine kinase receptor signaling, while LRRK2 is implicated in the pathogenesis of Parkinson's disease. In this report, we present a protocol to label the LRRK1 and LRRK2 proteins in cells with (32)P orthophosphate, thereby providing a means to measure the overall phosphorylation levels of these 2 proteins in cells. In brief, affinity tagged LRRK proteins are expressed in HEK293T cells which are exposed to medium containing (32)P-orthophosphate. The (32)P-orthophosphate is assimilated by the cells after only a few hours of incubation and all molecules in the cell containing phosphates are thereby radioactively labeled. Via the affinity tag (3xflag) the LRRK proteins are isolated from other cellular components by immunoprecipitation. Immunoprecipitates are then separated via SDS-PAGE, blotted to PVDF membranes and analysis of the incorporated phosphates is performed by autoradiography ((32)P signal) and western detection (protein signal) of the proteins on the blots. The protocol can readily be adapted to monitor phosphorylation of any other protein that can be expressed in cells and isolated by immunoprecipitation.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven and Leuven Institute for Neuroscience and Disease (LIND)
| | | | | |
Collapse
|
29
|
Wang Y, Liu W, He X, Zhou F. Parkinson's Disease-Associated Dj-1 Mutations Increase Abnormal Phosphorylation of Tau Protein through Akt/Gsk-3β Pathways. J Mol Neurosci 2013; 51:911-8. [DOI: 10.1007/s12031-013-0099-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022]
|
30
|
Rideout HJ, Stefanis L. The neurobiology of LRRK2 and its role in the pathogenesis of Parkinson's disease. Neurochem Res 2013; 39:576-92. [PMID: 23729298 DOI: 10.1007/s11064-013-1073-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/18/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large, widely expressed protein of largely unknown function. Mutations in the gene encoding LRRK2 have been linked to multiple diseases, including a prominent association with familial and sporadic Parkinson's disease (PD), as well as inflammatory bowel disorders such as Crohn's disease. The LRRK2 protein possesses both kinase and GTPase signaling domains, as well as multiple protein interaction domains. Experimental studies in both cellular and in vivo models of mutant LRRK2-induced neurodegeneration have given clues to potential function(s) of LRRK2, yet much remains unknown. For example, while it is known that intact kinase and GTPase activity are required for mutant forms of the protein to trigger cell death, the specific targets of these enzymatic activities that mediate the death of neurons are not known. In this review, we discuss the evidence linking LRRK2 to various cellular/neuronal activities such as extrinsic death and inflammatory signaling, lysosomal protein degradation, the cytoskeletal system and neurite outgrowth, vesicle trafficking, mitochondrial dysfunction, as well as multiple points of interaction with several other genes linked to the pathogenesis of PD. In order for more effective therapeutic strategies to be envisioned and implemented, the mechanisms underlying LRRK2-mediated neurodegeneration need to be better characterized. Furthermore, insights into LRRK2-associated PD pathogenesis can potentially advance our understanding of the more common sporadic forms of PD.
Collapse
Affiliation(s)
- Hardy J Rideout
- Laboratory of Neurodegenerative Diseases, Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece,
| | | |
Collapse
|
31
|
Berwick DC, Harvey K. LRRK2: an éminence grise of Wnt-mediated neurogenesis? Front Cell Neurosci 2013; 7:82. [PMID: 23754980 PMCID: PMC3668263 DOI: 10.3389/fncel.2013.00082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/13/2013] [Indexed: 01/13/2023] Open
Abstract
The importance of leucine-rich repeat kinase 2 (LRRK2) to mature neurons is well-established, since mutations in PARK8, the gene encoding LRRK2, are the most common known cause of Parkinson’s disease. Nonetheless, despite the LRRK2 knockout mouse having no overt neurodevelopmental defect, numerous lines of in vitro data point toward a central role for this protein in neurogenesis. Roles for LRRK2 have been described in many key processes, including neurite outgrowth and the regulation of microtubule dynamics. Moreover, LRRK2 has been implicated in cell cycle control, suggesting additional roles in neurogenesis that precede terminal differentiation. However, we contend that the suggested function of LRRK2 as a scaffolding protein at the heart of numerous Wnt signaling cascades provides the most tantalizing link to neurogenesis in the developing brain. Numerous lines of evidence show a critical requirement for multiple Wnt pathways in the development of certain brain regions, not least the dopaminergic neurons of the ventral mid-brain. In conclusion, these observations indicate a function of LRRK2 as a subtle yet critical mediator of the action of Wnt ligands on developing neurons. We suggest that LRRK2 loss- or gain-of-function are likely modifiers of developmental phenotypes seen in animal models of Wnt signaling deregulation, a hypothesis that can be tested by cross-breeding relevant genetically modified experimental strains.
Collapse
Affiliation(s)
- Daniel C Berwick
- Department of Pharmacology, University College London School of Pharmacy, University College London London, UK
| | | |
Collapse
|
32
|
Abstract
The PD (Parkinson's disease) protein LRRK2 (leucine-rich repeat kinase 2) occurs in cells as a highly phosphorylated protein, with the majority of phosphosites clustering in the region between the ankyrin repeat and leucine-rich repeat domains. The observation that several pathogenic variants of LRRK2 display strongly reduced cellular phosphorylation suggests that phosphorylation of LRRK2 is involved in the PD pathological process. Furthermore, treatment of cells with inhibitors of LRRK2 kinase activity, which are currently considered as potential disease-modifying therapeutics for PD, leads to a rapid decrease in the phosphorylation levels of LRRK2. For these reasons, understanding the cellular role and regulation of LRRK2 as a kinase and as a substrate has become the focus of intense investigation. In the present review, we discuss what is currently known about the cellular phosphorylation of LRRK2 and how this relates to its function and dysfunction.
Collapse
|
33
|
Abstract
LRRK2 (leucine-rich repeat kinase 2) is a large protein encoding multiple functional domains, including two catalytically active domains, a kinase and a GTPase domain. The LRRK2 GTPase belongs to the Ras-GTPase superfamily of GTPases, more specifically to the ROC (Ras of complex proteins) subfamily. Studies with recombinant LRRK2 protein purified from eukaryotic cells have confirmed that LRRK2 binds guanine nucleotides and catalyses the hydrolysis of GTP to GDP. LRRK2 is linked to PD (Parkinson's disease) and GTPase activity is impaired for several PD mutants located in the ROC and COR (C-terminal of ROC) domains, indicating that it is involved in PD pathogenesis. Ras family GTPases are known to function as molecular switches, and several studies have explored this possibility for LRRK2. These studies show that there is interplay between the LRRK2 GTPase function and its kinase function, with most data pointing towards a role for the kinase domain as an upstream regulator of ROC. The GTPase function is therefore a pivotal functionality within the LRRK2-mediated signalling cascade which includes partners encoded by other LRRK2 domains as well as other cellular signalling partners. The present review examines what is known of the enzymatic properties of the LRRK2 GTPase, the interplay between ROC and other LRRK2 domains, and the interplay between ROC and other cellular proteins with the dual goal to understand how LRRK2 GTPase affects cellular functions and point to future research venues.
Collapse
|
34
|
ATP-competitive LRRK2 inhibitors interfere with monoclonal antibody binding to the kinase domain of LRRK2 under native conditions. A method to directly monitor the active conformation of LRRK2? J Neurosci Methods 2013; 214:62-8. [PMID: 23318290 DOI: 10.1016/j.jneumeth.2012.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 11/21/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. LRRK2 kinase activity is required for toxicity in neuronal cell cultures suggesting that selective kinase inhibitors may prevent neurodegeneration in patients. Directly monitoring LRRK2 activity in cells would be advantageous for the development of small molecule LRRK2 inhibitors. Here, we demonstrate that a monoclonal anti-LRRK2 antibody directed against the activation segment binds less efficiently to native LRRK2 protein in the presence of ATP-competitive LRRK2 inhibitors. Since kinase inhibitors prevent autophosphorylation and refolding of the activation segment, we hypothesize that the antibody preferentially binds to the active conformation of LRRK2 under native conditions.
Collapse
|
35
|
Di Domenico F, Sultana R, Ferree A, Smith K, Barone E, Perluigi M, Coccia R, Pierce W, Cai J, Mancuso C, Squillace R, Wiengele M, Dalle-Donne I, Wolozin B, Butterfield DA. Redox proteomics analyses of the influence of co-expression of wild-type or mutated LRRK2 and Tau on C. elegans protein expression and oxidative modification: relevance to Parkinson disease. Antioxid Redox Signal 2012; 17:1490-506. [PMID: 22315971 PMCID: PMC3448940 DOI: 10.1089/ars.2011.4312] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/14/2012] [Accepted: 01/16/2012] [Indexed: 11/13/2022]
Abstract
AIMS The human LRRK2 gene has been identified as the most common causative gene of autosomal-dominantly inherited and idiopathic Parkinson disease (PD). The G2019S substitution is the most common mutation in LRRK2. The R1441C mutation also occurs in cases of familial PD, but is not as prevalent. Some cases of LRRK2-based PD exhibit Tau pathology, which suggests that alterations on LRRK2 activity affect the pathophysiology of Tau. To investigate how LRRK2 might affect Tau and the pathophysiology of PD, we generated lines of C. elegans expressing human LRRK2 [wild-type (WT) or mutated (G2019S or R1441C)] with and without V337M Tau. Expression and redox proteomics were used to identify the effects of LRRK2 (WT and mutant) on protein expression and oxidative modifications. RESULTS Co-expression of WT LRRK2 and Tau led to increased expression of numerous proteins, including several 60S ribosomal proteins, mitochondrial proteins, and the V-type proton ATPase, which is associated with autophagy. C. elegans expressing mutant LRRK2 showed similar changes, but also showed increased protein oxidation and lipid peroxidation, the latter indexed as increased protein-bound 4-hydroxy-2-nonenal (HNE). INNOVATION Our study brings new knowledge about the possible alterations induced by LRRK2 (WT and mutated) and Tau interactions, suggesting the involvement of G2019S and R1441C in Tau-dependent neurodegenerative processes. CONCLUSION These results suggest that changes in LRRK2 expression or activity lead to corresponding changes in mitochondrial function, autophagy, and protein translation. These findings are discussed with reference to the pathophysiology of PD.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Andrew Ferree
- Department of Pharmacology, Boston University, Boston, Massachusetts
| | - Katelyn Smith
- Department of Pharmacology, Boston University, Boston, Massachusetts
| | - Eugenio Barone
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
- Institute of Pharmacology, Catholic University of Rome, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Raffaella Coccia
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - William Pierce
- Department of Pharmacology, University of Louisville, Louisville, Kentucky
| | - Jian Cai
- Department of Pharmacology, University of Louisville, Louisville, Kentucky
| | - Cesare Mancuso
- Institute of Pharmacology, Catholic University of Rome, Rome, Italy
| | | | | | | | - Benjamin Wolozin
- Department of Pharmacology, Boston University, Boston, Massachusetts
- Edith Nourse Rogers Memorial Veterans Affairs Medical Center, Bedford, Massachusetts
| | | |
Collapse
|
36
|
Löw K, Aebischer P. Use of viral vectors to create animal models for Parkinson's disease. Neurobiol Dis 2012; 48:189-201. [DOI: 10.1016/j.nbd.2011.12.038] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 12/20/2011] [Indexed: 12/15/2022] Open
|
37
|
Belluzzi E, Greggio E, Piccoli G. Presynaptic dysfunction in Parkinson's disease: a focus on LRRK2. Biochem Soc Trans 2012; 40:1111-1116. [PMID: 22988874 DOI: 10.1042/bst20120124] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
PD (Parkinson's disease) is a common neurodegenerative disease clinically characterized by bradykinesia, rigidity and resting tremor. Recent studies have proposed that synaptic dysfunction, implicated in numerous studies of animal models of PD, might be a key factor in PD. The molecular defects that lead to PD progression might be hidden at the presynaptic neuron: in fact accumulating evidence has shown that the majority of the genes linked to PD play a critical role at the presynaptic site. In the present paper, we focus on the presynaptic function of LRRK2 (leucine-rich repeat kinase 2), a protein that mutated represents the main genetic cause of familial PD described to date. Neurotransmission relies on proper presynaptic vesicle trafficking; defects in this process, variation in dopamine flow and alteration of presynaptic plasticity have been reported in several animal models of LRRK2 mutations. Furthermore, impaired dopamine turnover has been described in presymptomatic LRRK2 PD patients. Thus, given the pathological events occurring at the synapses of PD patients, the presynaptic site may represent a promising target for early diagnostic therapeutic intervention.
Collapse
Affiliation(s)
- Elisa Belluzzi
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, Padua, Italy
| | | | | |
Collapse
|
38
|
Civiero L, Vancraenenbroeck R, Belluzzi E, Beilina A, Lobbestael E, Reyniers L, Gao F, Micetic I, De Maeyer M, Bubacco L, Baekelandt V, Cookson MR, Greggio E, Taymans JM. Biochemical characterization of highly purified leucine-rich repeat kinases 1 and 2 demonstrates formation of homodimers. PLoS One 2012; 7:e43472. [PMID: 22952686 PMCID: PMC3430690 DOI: 10.1371/journal.pone.0043472] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/20/2012] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeat kinase 1 and 2 (LRRK1 and LRRK2) are large multidomain proteins containing kinase, GTPase and multiple protein-protein interaction domains, but only mutations in LRRK2 are linked to familial Parkinson's disease (PD). Independent studies suggest that LRRK2 exists in the cell as a complex compatible with the size of a dimer. However, whether this complex is truly a homodimer or a heterologous complex formed by monomeric LRRK2 with other proteins has not been definitively proven due to the limitations in obtaining highly pure proteins suitable for structural characterization. Here, we used stable expression of LRRK1 and LRRK2 in HEK293T cell lines to produce recombinant LRRK1 and LRRK2 proteins of greater than 90% purity. Both purified LRRKs are folded, with a predominantly alpha-helical secondary structure and are capable of binding GTP with similar affinity. Furthermore, recombinant LRRK2 exhibits robust autophosphorylation activity, phosphorylation of model peptides in vitro and ATP binding. In contrast, LRRK1 does not display significant autophosphorylation activity and fails to phosphorylate LRRK2 model substrates, although it does bind ATP. Using these biochemically validated proteins, we show that LRRK1 and LRRK2 are capable of forming homodimers as shown by single-particle transmission electron microscopy and immunogold labeling. These LRRK dimers display an elongated conformation with a mean particle size of 145 Å and 175 Å respectively, which is disrupted by addition of 6M guanidinium chloride. Immunogold staining revealed double-labeled particles also in the pathological LRRK2 mutant G2019S and artificial mutants disrupting GTPase and kinase activities, suggesting that point mutations do not hinder the dimeric conformation. Overall, our findings indicate for the first time that purified and active LRRK1 and LRRK2 can form dimers in their full-length conformation.
Collapse
Affiliation(s)
- Laura Civiero
- Department of Biology, University of Padova, Padova, Italy
| | - Renée Vancraenenbroeck
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
- Laboratory for Biomolecular Modelling, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Elisa Belluzzi
- Department of Biology, University of Padova, Padova, Italy
| | - Alexandra Beilina
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lauran Reyniers
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Fangye Gao
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ivan Micetic
- Department of Biology, University of Padova, Padova, Italy
| | - Marc De Maeyer
- Laboratory for Biomolecular Modelling, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, Italy
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mark R. Cookson
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
- * E-mail: (EG); (JMT)
| | - Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium
- * E-mail: (EG); (JMT)
| |
Collapse
|
39
|
Wang C, Cai Y, Zheng Z, Tang BS, Xu Y, Wang T, Ma J, Chen SD, Langston JW, Tanner CM, Chan P. Penetrance of LRRK2 G2385R and R1628P is modified by common PD-associated genetic variants. Parkinsonism Relat Disord 2012; 18:958-63. [PMID: 22658533 DOI: 10.1016/j.parkreldis.2012.05.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 05/02/2012] [Accepted: 05/03/2012] [Indexed: 01/02/2023]
Abstract
Variants in the LRRK2 gene are well-characterized genetic predisposing factors for PD worldwide, and LRRK2-associated PD is often indistinguishable from idiopathic PD (IPD). However, considerable heterogeneity of LRRK2-PD suggests the existence of additional genetic and/or environmental modifiers for LRRK2 carriers, which have yet to be confirmed by large-scale human studies. In a Chinese cohort consisting of 2013 sporadic PD patients and 1971 controls, we investigated the modification of the two Asian-specific LRRK2 variants, G2385R and R1628P, by variants of five other PD-associated genes/loci (SNCA, MAPT, GBA, BST1, PARK16). Of all the PD patients, 13.1% carried LRRK2 G2385R and/or R1628P variant. Among these carriers, a total of 15 different polygenic genotypes were detected representing different combination patterns between LRRK2 variants and those of the other genes/loci, which, alone or in combination, significantly modified the LRRK2-related risk for PD and the patients' ages at onset (AAOs). These results not only represent the largest replication data affirming the association between PD and all the six genes/loci in Chinese, but for the first time suggest that multiple PD-associated genetic factors modify both the penetrance and AAO of LRRK2 parkinsonism. This finding may have important implications for elucidating pathophysiologic mechanisms relevant to both LRRK2-associated and idiopathic PD. However, testing interactions among multiple genes by genetic association studies is still challenging. Future studies with much larger sample sizes are needed to confirm our findings.
Collapse
Affiliation(s)
- Chaodong Wang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, #45Changchun Street, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kinase inhibitors for CNS diseases: an analysis of the recent patent literature. Pharm Pat Anal 2012; 1:177-92. [DOI: 10.4155/ppa.12.19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein kinases (PKs), as members of an important target class in current pharmaceutical research, have been mostly exploited so far in therapeutic areas such as oncology and inflammation. However, basic research on some PKs as key components of molecular mechanisms underlying neurodegeneration and neuroprotection may translate into new medicines for CNS diseases in the next few years. This review is an account of recent patents dealing with kinase inhibitors primarily designed for CNS indications. CNS-directed patents on kinase modulators published after 2008 were surveyed using SciFinder® and public patent search engines. Some PK targets, such as GSK-3β, CDK5, ROCK and p38α MAPK, continue to attract interest even though a clinical proof-of-concept is yet to be attained in a CNS setting. Less established PKs such as LRRK2, MLK, PAK and DAPK-1 hold promise as valuable targets of the future.
Collapse
|
41
|
Ujiie S, Hatano T, Kubo SI, Imai S, Sato S, Uchihara T, Yagishita S, Hasegawa K, Kowa H, Sakai F, Hattori N. LRRK2 I2020T mutation is associated with tau pathology. Parkinsonism Relat Disord 2012; 18:819-23. [PMID: 22525366 DOI: 10.1016/j.parkreldis.2012.03.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 03/08/2012] [Accepted: 03/21/2012] [Indexed: 10/28/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of autosomal-dominant familial Parkinson's disease (FPD). The variable pathological features of LRRK2-linked FPD include Lewy bodies, degeneration of anterior horn cells associated with axonal spheroids, neurofibrillary tangles (NFTs) and TAR DNA-binding protein of 43 kDa (TDP-43) positive inclusion bodies. Furthermore, abnormal hyperphosphorylation of microtubule associated protein tau, in part generated by catalysis of protein kinases, has been reported to be involved in progressive neurodegeneration in a number of diseases, including FPD. Thus, we examined six patients carrying the LRRK2 I2020T mutation, a pathogenic mutation associated with PARK8, and found abnormal tau phosphorylation depositions in the brainstem. Additionally, we found LRRK2 I2020T enhanced tau phosphorylation in cultured cells co-expressing LRRK2-I2020T and 3 or 4-repeated tau. This is the first report describing the relationship between hyperphosphorylation of tau and LRRK2 I2020T.
Collapse
Affiliation(s)
- Sachiko Ujiie
- Department of Neurology, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Expression, purification and preliminary biochemical and structural characterization of the leucine rich repeat namesake domain of leucine rich repeat kinase 2. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1824:450-60. [DOI: 10.1016/j.bbapap.2011.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 12/10/2011] [Accepted: 12/27/2011] [Indexed: 01/13/2023]
|
43
|
Synaptic Dysfunction in Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:553-72. [DOI: 10.1007/978-3-7091-0932-8_24] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
44
|
Bardien S, Lesage S, Brice A, Carr J. Genetic characteristics of leucine-rich repeat kinase 2 (LRRK2) associated Parkinson's disease. Parkinsonism Relat Disord 2011; 17:501-8. [PMID: 21641266 DOI: 10.1016/j.parkreldis.2010.11.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 11/09/2010] [Accepted: 11/09/2010] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive and selective degeneration of nigrostriatal dopaminergic neurons. The discovery of at least six PD-causing genes in predominantly early-onset forms of the disorder has cemented a genetic component to the etiology. Notably, the discovery of mutations in the LRRK2 gene in patients presenting with typical 'sporadic' PD with ages at onset in their sixties and seventies has shifted paradigms in the field of PD research. The G2019S mutation in LRRK2 has been found in diverse populations worldwide and usually resides on a common haplotype revealing that many of these individuals share a common ancestor, probably of Middle Eastern origin. The only validated coding susceptibility alleles for PD, G2385R and R1628P, are both in this gene but to date have been found exclusively in Asian populations. Concomitant with genetic testing for PD is the need for appropriate and informed genetic counseling. Families of patients with LRRK2 mutations and susceptibility alleles need to be informed about the current lack of disease preventative strategies and the implications surrounding incomplete penetrance. In summary, single-handedly LRRK2 has had a major impact on the field of PD research and the findings have been of interest to both clinicians and scientists. We anticipate that other genes of such major impact exist for PD and look forward to their discovery.
Collapse
Affiliation(s)
- Soraya Bardien
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| | | | | | | |
Collapse
|
45
|
Doggett EA, Zhao J, Mork CN, Hu D, Nichols RJ. Phosphorylation of LRRK2 serines 955 and 973 is disrupted by Parkinson's disease mutations and LRRK2 pharmacological inhibition. J Neurochem 2011; 120:37-45. [PMID: 22004453 DOI: 10.1111/j.1471-4159.2011.07537.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease. An amino terminal cluster of constitutively phosphorylated residues, serines 860, 910, 935, 955, and 973, appears to be biologically relevant. Phosphorylation of serines 910 and 935 is regulated in response to LRRK2 kinase activity and is responsible for interaction with 14-3-3 and maintaining LRRK2 in a non-aggregated state. We examined the phosphorylation status of two other constitutive phosphorylation sites, serines 955 and 973. Treatment of LRRK2 expressing cells with the selective LRRK2 inhibitor LRRK2-IN1 revealed that, like Ser910/Ser935, phosphorylation of Ser955 and Ser973 is disrupted by acute inhibition of LRRK2 kinase activity. Additionally, phosphorylation of Ser955 and 973 is disrupted in the context of several Parkinson's disease associated mutations [R1441G/C, Y1699C, and I2020T]. We observed that modification of Ser973 is dependent on the modification of Ser910/Ser935. Ser955Ala and Ser973Ala mutations do not induce relocalization of LRRK2; however, all phosphomutants exhibited similar localization patterns when exposed to LRRK2-IN1. We conclude that the mechanisms of regulation of Ser910/935/955/973 phosphorylation are similar and physiologically relevant. These sites can be utilized as biomarkers for LRRK2 activity as well as starting points for the elucidation of upstream and downstream enzymes that regulate LRRK2.
Collapse
|
46
|
Taymans JM, Vancraenenbroeck R, Ollikainen P, Beilina A, Lobbestael E, De Maeyer M, Baekelandt V, Cookson MR. LRRK2 kinase activity is dependent on LRRK2 GTP binding capacity but independent of LRRK2 GTP binding. PLoS One 2011; 6:e23207. [PMID: 21858031 PMCID: PMC3155532 DOI: 10.1371/journal.pone.0023207] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/12/2011] [Indexed: 11/18/2022] Open
Abstract
Leucine rich repeat kinase 2 (LRRK2) is a Parkinson's disease (PD) gene that encodes a large multidomain protein including both a GTPase and a kinase domain. GTPases often regulate kinases within signal transduction cascades, where GTPases act as molecular switches cycling between a GTP bound “on” state and a GDP bound “off” state. It has been proposed that LRRK2 kinase activity may be increased upon GTP binding at the LRRK2 Ras of complex proteins (ROC) GTPase domain. Here we extensively test this hypothesis by measuring LRRK2 phosphorylation activity under influence of GDP, GTP or non-hydrolyzable GTP analogues GTPγS or GMPPCP. We show that autophosphorylation and lrrktide phosphorylation activity of recombinant LRRK2 protein is unaltered by guanine nucleotides, when co-incubated with LRRK2 during phosphorylation reactions. Also phosphorylation activity of LRRK2 is unchanged when the LRRK2 guanine nucleotide binding pocket is previously saturated with various nucleotides, in contrast to the greatly reduced activity measured for the guanine nucleotide binding site mutant T1348N. Interestingly, when nucleotides were incubated with cell lysates prior to purification of LRRK2, kinase activity was slightly enhanced by GTPγS or GMPPCP compared to GDP, pointing to an upstream guanine nucleotide binding protein that may activate LRRK2 in a GTP-dependent manner. Using metabolic labeling, we also found that cellular phosphorylation of LRRK2 was not significantly modulated by nucleotides, although labeling is significantly reduced by guanine nucleotide binding site mutants. We conclude that while kinase activity of LRRK2 requires an intact ROC-GTPase domain, it is independent of GDP or GTP binding to ROC.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Laboratory for Neurobiology and Gene Therapy, Katholieke Universiteit Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Biernacka JM, Armasu SM, Cunningham JM, Ahlskog JE, Chung SJ, Maraganore DM. Do interactions between SNCA, MAPT, and LRRK2 genes contribute to Parkinson's disease susceptibility? Parkinsonism Relat Disord 2011; 17:730-6. [PMID: 21816655 DOI: 10.1016/j.parkreldis.2011.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 06/21/2011] [Accepted: 07/01/2011] [Indexed: 11/26/2022]
Abstract
BACKGROUND Polymorphisms in SNCA, MAPT and LRRK2 genes have recently been confirmed as risk factors for Parkinson's disease (PD), although with small individual attributable risk. Here we investigated the association of PD with interactions between variants of these genes. METHODS As part of a previous study of PD susceptibility genes 119 SNCA, MAPT, and LRRK2 haplotype tagging single nucleotide polymorphisms (SNPs) and two variable number tandem repeats (VNTRs) were genotyped in 1098 PD cases from the upper Midwest, USA and 1098 matched controls. Twenty-six of these SNPs were selected for SNP-SNP (or SNP-VNTR or VNTR-VNTR) interaction analysis (256 interaction pairs). Case-control analyses were performed to study association of pairwise SNP interactions with PD susceptibility. RESULTS Out of the 256 interaction pairs investigated, 10 had uncorrected p-values <0.05. These represented six SNCA-LRRK2 pairs, three SNCA-MAPT pairs, and one MAPT-LRRK2 pair. However, none of these pairwise interactions were significant after correction for multiple testing. Secondary analyses in strata defined by type of control (sibling or unrelated), sex, or age at onset of the case also did not reveal any significant interactions after accounting for multiple testing. CONCLUSIONS This study provides no statistically significant evidence of gene-gene interaction effects for the three confirmed genetic susceptibility loci for PD. However, this does not exclude the possibility that other genomic loci or environmental risk factors interact with these genes.
Collapse
Affiliation(s)
- Joanna M Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55095, USA
| | | | | | | | | | | |
Collapse
|
48
|
Plowey ED, Chu CT. Synaptic dysfunction in genetic models of Parkinson's disease: a role for autophagy? Neurobiol Dis 2011; 43:60-7. [PMID: 20969957 PMCID: PMC3049988 DOI: 10.1016/j.nbd.2010.10.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 10/08/2010] [Accepted: 10/13/2010] [Indexed: 11/21/2022] Open
Abstract
The past decade in Parkinson's disease (PD) research has been punctuated by numerous advances in understanding genetic factors that contribute to the disease. Common to most of the genetic models of Parkinsonian neurodegeneration are pathologic mechanisms of mitochondrial dysfunction, secretory vesicle dysfunction and oxidative stress that likely trigger common cell death mechanisms. Whereas presynaptic function is implicated in the function/dysfunction of α-synuclein, the first gene shown to contribute to PD, synaptic function has not comprised a major focus in most other genetic models. However, recent advances in understanding the impact of mutations in parkin and LRRK2 have also yielded insights into synaptic dysfunction as a possible early pathogenic mechanism. Autophagy is a common neuronal response in each of these genetic models of PD, participating in the clearance of protein aggregates and injured mitochondria. However, the potential consequences of autophagy upregulation on synaptic structure and function remain unknown. In this review, we discuss the evidence that supports a role for synaptic dysfunction in the neurodegenerative cascade in PD, and highlight unresolved questions concerning a potential role for autophagy in either pathological or compensatory synaptic remodeling. This article is part of a Special Issue entitled "Autophagy and protein degradation in neurological diseases."
Collapse
Affiliation(s)
- Edward D Plowey
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
49
|
Abstract
Parkinson disease is a common and usually sporadic neurodegenerative disorder. However, a subset of cases are inherited and, of these, mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are the most frequent genetic cause of disease. Here, we will discuss recent progress in understanding how LRRK2 mutations lead to disease and how this might have therapeutic implications. The effect of mutations on LRRK2 enzyme function provides clues as to which functions of the protein are important to disease. Recent work has focused on the kinase and GTP-binding domains of LRRK2, and it is assumed that these will be therapeutically important, although there is a substantial amount of work to be done to address this hypothesis.
Collapse
Affiliation(s)
- Azad Kumar
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892-3707, USA
| | | |
Collapse
|
50
|
Berwick DC, Harvey K. LRRK2 signaling pathways: the key to unlocking neurodegeneration? Trends Cell Biol 2011; 21:257-65. [PMID: 21306901 DOI: 10.1016/j.tcb.2011.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 12/21/2010] [Accepted: 01/04/2011] [Indexed: 11/16/2022]
Abstract
Mutations in PARK8, encoding leucine-rich repeat kinase 2 (LRRK2), are a major cause of Parkinson's disease. We contrast data suggesting that changes in LRRK2 activity cause alterations in mitogen-activated protein kinase, translational control, tumor necrosis factor α/Fas ligand and Wnt signaling pathways with the cell biological functions of LRRK2 such as vesicle trafficking. Despite scarce in vivo data on cell signaling, involvement in diverse cell biological functions suggests a role for LRRK2 as an upstream regulator in events leading to neurodegeneration. To stimulate discussion and give direction for future research, we further suggest that despite the importance of the catalytic activity for cytotoxicity, the main cellular function of LRRK2 is linked to assembly of signaling complexes.
Collapse
Affiliation(s)
- Daniel C Berwick
- Department of Pharmacology, School of Pharmacy, University of London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | | |
Collapse
|