1
|
Bayati A, McPherson PS. Alpha-synuclein, autophagy-lysosomal pathway, and Lewy bodies: Mutations, propagation, aggregation, and the formation of inclusions. J Biol Chem 2024; 300:107742. [PMID: 39233232 PMCID: PMC11460475 DOI: 10.1016/j.jbc.2024.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
Research into the pathophysiology of Parkinson's disease (PD) is a fast-paced pursuit, with new findings about PD and other synucleinopathies being made each year. The involvement of various lysosomal proteins, such as TFEB, TMEM175, GBA, and LAMP1/2, marks the rising awareness about the importance of lysosomes in PD and other neurodegenerative disorders. This, along with recent developments regarding the involvement of microglia and the immune system in neurodegenerative diseases, has brought about a new era in neurodegeneration: the role of proinflammatory cytokines on the nervous system, and their downstream effects on mitochondria, lysosomal degradation, and autophagy. More effort is needed to understand the interplay between neuroimmunology and disease mechanisms, as many of the mechanisms remain enigmatic. α-synuclein, a key protein in PD and the main component of Lewy bodies, sits at the nexus between lysosomal degradation, autophagy, cellular stress, neuroimmunology, PD pathophysiology, and disease progression. This review revisits some fundamental knowledge about PD while capturing some of the latest trends in PD research, specifically as it relates to α-synuclein.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Post-translational glycosylation diminishes α-synuclein pathology formation. Nat Chem Biol 2024; 20:553-554. [PMID: 38355724 DOI: 10.1038/s41589-024-01553-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
|
3
|
Garmendia JV, De Sanctis CV, Das V, Annadurai N, Hajduch M, De Sanctis JB. Inflammation, Autoimmunity and Neurodegenerative Diseases, Therapeutics and Beyond. Curr Neuropharmacol 2024; 22:1080-1109. [PMID: 37898823 PMCID: PMC10964103 DOI: 10.2174/1570159x22666231017141636] [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: 06/05/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 10/30/2023] Open
Abstract
Neurodegenerative disease (ND) incidence has recently increased due to improved life expectancy. Alzheimer's (AD) or Parkinson's disease (PD) are the most prevalent NDs. Both diseases are poly genetic, multifactorial and heterogenous. Preventive medicine, a healthy diet, exercise, and controlling comorbidities may delay the onset. After the diseases are diagnosed, therapy is needed to slow progression. Recent studies show that local, peripheral and age-related inflammation accelerates NDs' onset and progression. Patients with autoimmune disorders like inflammatory bowel disease (IBD) could be at higher risk of developing AD or PD. However, no increase in ND incidence has been reported if the patients are adequately diagnosed and treated. Autoantibodies against abnormal tau, β amyloid and α- synuclein have been encountered in AD and PD and may be protective. This discovery led to the proposal of immune-based therapies for AD and PD involving monoclonal antibodies, immunization/ vaccines, pro-inflammatory cytokine inhibition and anti-inflammatory cytokine addition. All the different approaches have been analysed here. Future perspectives on new therapeutic strategies for both disorders are concisely examined.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Marián Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| |
Collapse
|
4
|
Chocarro J, Rico AJ, Ariznabarreta G, Roda E, Honrubia A, Collantes M, Peñuelas I, Vázquez A, Rodríguez-Pérez AI, Labandeira-García JL, Vila M, Lanciego JL. Neuromelanin accumulation drives endogenous synucleinopathy in non-human primates. Brain 2023; 146:5000-5014. [PMID: 37769648 PMCID: PMC10689915 DOI: 10.1093/brain/awad331] [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: 04/19/2023] [Revised: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023] Open
Abstract
Although neuromelanin is a dark pigment characteristic of dopaminergic neurons in the human substantia nigra pars compacta, its potential role in the pathogenesis of Parkinson's disease (PD) has often been neglected since most commonly used laboratory animals lack neuromelanin. Here we took advantage of adeno-associated viral vectors encoding the human tyrosinase gene for triggering a time-dependent neuromelanin accumulation within substantia nigra pars compacta dopaminergic neurons in macaques up to similar levels of pigmentation as observed in elderly humans. Furthermore, neuromelanin accumulation induced an endogenous synucleinopathy mimicking intracellular inclusions typically observed in PD together with a progressive degeneration of neuromelanin-expressing dopaminergic neurons. Moreover, Lewy body-like intracellular inclusions were observed in cortical areas of the frontal lobe receiving dopaminergic innervation, supporting a circuit-specific anterograde spread of endogenous synucleinopathy by permissive trans-synaptic templating. In summary, the conducted strategy resulted in the development and characterization of a new macaque model of PD matching the known neuropathology of this disorder with unprecedented accuracy. Most importantly, evidence is provided showing that intracellular aggregation of endogenous α-synuclein is triggered by neuromelanin accumulation, therefore any therapeutic approach intended to decrease neuromelanin levels may provide appealing choices for the successful implementation of novel PD therapeutics.
Collapse
Affiliation(s)
- Julia Chocarro
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Alberto J Rico
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Goiaz Ariznabarreta
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Elvira Roda
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Adriana Honrubia
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - María Collantes
- Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Iván Peñuelas
- Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alfonso Vázquez
- Department of Neurosurgery, Hospital Universitario de Navarra, Servicio Navarro de Salud, 31008 Pamplona, Spain
| | - Ana I Rodríguez-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José L Labandeira-García
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Miquel Vila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Vall d’Hebron Research Institute, Neurodegenerative Diseses Research Group, 08035 Barcelona, Spain
- Autonomous University of Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - José L Lanciego
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
5
|
Heid LF, Kupreichyk T, Schützmann MP, Schneider W, Stoldt M, Hoyer W. Nucleation of α-Synuclein Amyloid Fibrils Induced by Cross-Interaction with β-Hairpin Peptides Derived from Immunoglobulin Light Chains. Int J Mol Sci 2023; 24:16132. [PMID: 38003322 PMCID: PMC10671648 DOI: 10.3390/ijms242216132] [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: 09/15/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Heterologous interactions between different amyloid-forming proteins, also called cross-interactions, may have a critical impact on disease-related amyloid formation. β-hairpin conformers of amyloid-forming proteins have been shown to affect homologous interactions in the amyloid self-assembly process. Here, we applied two β-hairpin-forming peptides derived from immunoglobulin light chains as models to test how heterologous β-hairpins modulate the fibril formation of Parkinson's disease-associated protein α-synuclein (αSyn). The peptides SMAhp and LENhp comprise β-strands C and C' of the κ4 antibodies SMA and LEN, which are associated with light chain amyloidosis and multiple myeloma, respectively. SMAhp and LENhp bind with high affinity to the β-hairpin-binding protein β-wrapin AS10 according to isothermal titration calorimetry and NMR spectroscopy. The addition of SMAhp and LENhp affects the kinetics of αSyn aggregation monitored by Thioflavin T (ThT) fluorescence, with the effect depending on assay conditions, salt concentration, and the applied β-hairpin peptide. In the absence of agitation, substoichiometric concentrations of the hairpin peptides strongly reduce the lag time of αSyn aggregation, suggesting that they support the nucleation of αSyn amyloid fibrils. The effect is also observed for the aggregation of αSyn fragments lacking the N-terminus or the C-terminus, indicating that the promotion of nucleation involves the interaction of hairpin peptides with the hydrophobic non-amyloid-β component (NAC) region.
Collapse
Affiliation(s)
- Laetitia F. Heid
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Tatsiana Kupreichyk
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Marie P. Schützmann
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Walfried Schneider
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Matthias Stoldt
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
6
|
Xiang J, Tao Y, Xia Y, Luo S, Zhao Q, Li B, Zhang X, Sun Y, Xia W, Zhang M, Kang SS, Ahn EH, Liu X, Xie F, Guan Y, Yang JJ, Bu L, Wu S, Wang X, Cao X, Liu C, Zhang Z, Li D, Ye K. Development of an α-synuclein positron emission tomography tracer for imaging synucleinopathies. Cell 2023; 186:3350-3367.e19. [PMID: 37421950 PMCID: PMC10527432 DOI: 10.1016/j.cell.2023.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 07/10/2023]
Abstract
Synucleinopathies are characterized by the accumulation of α-synuclein (α-Syn) aggregates in the brain. Positron emission tomography (PET) imaging of synucleinopathies requires radiopharmaceuticals that selectively bind α-Syn deposits. We report the identification of a brain permeable and rapid washout PET tracer [18F]-F0502B, which shows high binding affinity for α-Syn, but not for Aβ or Tau fibrils, and preferential binding to α-Syn aggregates in the brain sections. Employing several cycles of counter screenings with in vitro fibrils, intraneuronal aggregates, and neurodegenerative disease brain sections from several mice models and human subjects, [18F]-F0502B images α-Syn deposits in the brains of mouse and non-human primate PD models. We further determined the atomic structure of the α-Syn fibril-F0502B complex by cryo-EM and revealed parallel diagonal stacking of F0502B on the fibril surface through an intense noncovalent bonding network via inter-ligand interactions. Therefore, [18F]-F0502B is a promising lead compound for imaging aggregated α-Syn in synucleinopathies.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Biomedical Sciences, School of Medicine, JiangHan University, #8, Sanjiaohu Rd., Wuhan 430056, China
| | - Shilin Luo
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Yunpeng Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Mingming Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eun-Hee Ahn
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Lihong Bu
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shengxi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
7
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
8
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
9
|
Chu Y, Hirst WD, Kordower JH. Mixed pathology as a rule, not exception: Time to reconsider disease nosology. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:57-71. [PMID: 36796948 DOI: 10.1016/b978-0-323-85538-9.00012-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder that is associated with motor and nonmotor symptoms. Accumulation of misfolded α-synuclein is considered a key pathological feature during disease initiation and progression. While clearly deemed a synucleinopathy, the development of amyloid-β plaques, tau-containing neurofibrillary tangles, and even TDP-43 protein inclusions occur within the nigrostriatal system and in other brain regions. In addition, inflammatory responses, manifested by glial reactivity, T-cell infiltration, and increased expression of inflammatory cytokines, plus other toxic mediators derived from activated glial cells, are currently recognized as prominent drivers of Parkinson's disease pathology. However, copathologies have increasingly been recognized as the rule (>90%) and not the exception, with Parkinson's disease cases on average exhibiting three different copathologies. While microinfarcts, atherosclerosis, arteriolosclerosis, and cerebral amyloid angiopathy may have an impact on disease progression, α-synuclein, amyloid-β, and TDP-43 pathology do not seem to contribute to progression.
Collapse
Affiliation(s)
- Yaping Chu
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Boston, MA, United States
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
10
|
Xie L, Hu L. Research progress in the early diagnosis of Parkinson’s disease. Neurol Sci 2022; 43:6225-6231. [DOI: 10.1007/s10072-022-06316-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
|
11
|
Lang AE, Siderowf AD, Macklin EA, Poewe W, Brooks DJ, Fernandez HH, Rascol O, Giladi N, Stocchi F, Tanner CM, Postuma RB, Simon DK, Tolosa E, Mollenhauer B, Cedarbaum JM, Fraser K, Xiao J, Evans KC, Graham DL, Sapir I, Inra J, Hutchison RM, Yang M, Fox T, Budd Haeberlein S, Dam T. Trial of Cinpanemab in Early Parkinson's Disease. N Engl J Med 2022; 387:408-420. [PMID: 35921450 DOI: 10.1056/nejmoa2203395] [Citation(s) in RCA: 145] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Aggregated α-synuclein plays an important role in Parkinson's disease pathogenesis. Cinpanemab, a human-derived monoclonal antibody that binds to α-synuclein, is being evaluated as a disease-modifying treatment for Parkinson's disease. METHODS In a 52-week, multicenter, double-blind, phase 2 trial, we randomly assigned, in a 2:1:2:2 ratio, participants with early Parkinson's disease to receive intravenous infusions of placebo (control) or cinpanemab at a dose of 250 mg, 1250 mg, or 3500 mg every 4 weeks, followed by an active-treatment dose-blinded extension period for up to 112 weeks. The primary end points were the changes from baseline in the Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) total score (range, 0 to 236, with higher scores indicating worse performance) at weeks 52 and 72. Secondary end points included MDS-UPDRS subscale scores and striatal binding as assessed on dopamine transporter single-photon-emission computed tomography (DaT-SPECT). RESULTS Of the 357 enrolled participants, 100 were assigned to the control group, 55 to the 250-mg cinpanemab group, 102 to the 1250-mg group, and 100 to the 3500-mg group. The trial was stopped after the week 72 interim analysis owing to lack of efficacy. The change to week 52 in the MDS-UPDRS score was 10.8 points in the control group, 10.5 points in the 250-mg group, 11.3 points in the 1250-mg group, and 10.9 points in the 3500-mg group (adjusted mean difference vs. control, -0.3 points [95% confidence interval {CI}, -4.9 to 4.3], P = 0.90; 0.5 points [95% CI, -3.3 to 4.3], P = 0.80; and 0.1 point [95% CI, -3.8 to 4.0], P = 0.97, respectively). The adjusted mean difference at 72 weeks between participants who received cinpanemab through 72 weeks and the pooled group of those who started cinpanemab at 52 weeks was -0.9 points (95% CI, -5.6 to 3.8) for the 250-mg dose, 0.6 points (95% CI, -3.3 to 4.4) for the 1250-mg dose, and -0.8 points (95% CI, -4.6 to 3.0) for the 3500-mg dose. Results for secondary end points were similar to those for the primary end points. DaT-SPECT imaging at week 52 showed no differences between the control group and any cinpanemab group. The most common adverse events with cinpanemab were headache, nasopharyngitis, and falls. CONCLUSIONS In participants with early Parkinson's disease, the effects of cinpanemab on clinical measures of disease progression and changes in DaT-SPECT imaging did not differ from those of placebo over a 52-week period. (Funded by Biogen; SPARK ClinicalTrials.gov number, NCT03318523.).
Collapse
Affiliation(s)
- Anthony E Lang
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Andrew D Siderowf
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Eric A Macklin
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Werner Poewe
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - David J Brooks
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Hubert H Fernandez
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Olivier Rascol
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Nir Giladi
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Fabrizio Stocchi
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Caroline M Tanner
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Ronald B Postuma
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - David K Simon
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Eduardo Tolosa
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Brit Mollenhauer
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Jesse M Cedarbaum
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Kyle Fraser
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - James Xiao
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Karleyton C Evans
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Danielle L Graham
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Inbal Sapir
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Jennifer Inra
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - R Matthew Hutchison
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Minhua Yang
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Tara Fox
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Samantha Budd Haeberlein
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| | - Tien Dam
- From the Edmond J. Safra Program in Parkinson's Disease, University Health Network, and the University of Toronto, Toronto (A.E.L.), and the Montreal Neurological Institute, Montreal (R.B.P.); the University of Pennsylvania, Philadelphia (A.D.S.); the Biostatistics Center, Massachusetts General Hospital (E.A.M.), Beth Israel Deaconess Medical Center (D.K.S.), and Harvard Medical School (E.A.M., D.K.S.), Boston, and Biogen, Cambridge (K.F., J.X., K.C.E., D.L.G., I.S., J.I., R.M.H., M.Y., S.B.H., T.D.) - all in Massachusetts; Medizinische Universität Innsbruck, Innsbruck, Austria (W.P.); Newcastle University, Newcastle upon Tyne (D.J.B.), and Biogen, Maidenhead (T.F.) - both in the United Kingdom; Aarhus University, Aarhus, Denmark (D.J.B.); the Center for Neurological Restoration, Cleveland Clinic, and Cleveland Clinic Lerner College of Medicine - both in Cleveland (H.H.F.); Clinical Investigation Center 1436, the Departments of Clinical Pharmacology and Neurosciences, NS-PARK-French Clinical Research Infrastructure Network, NeuroToul COEN Center, INSERM, University Hospital of Toulouse, and the University of Toulouse III - both in Toulouse, France (O.R.); Tel Aviv Sourasky Medical Center, and the Sackler School of Medicine and the Sagol School of Neuroscience, Tel Aviv University - both in Tel Aviv, Israel (N.G.); University San Raffaele and IRCCS San Raffaele - both in Rome (F.S.); the University of California, San Diego, La Jolla (C.M.T.), and the San Francisco Veterans Affairs Medical Center, San Francisco (C.M.T.); the University of Barcelona, Barcelona (E.T.); the Department of Neurology, University Medical Center Göttingen, Göttingen, and Paracelsus-Elena-Klinik, Kassel - both in Germany (B.M.); and Coeruleus Clinical Sciences, Woodbridge, CT (J.M.C.)
| |
Collapse
|
12
|
Moudio S, Rodin F, Albargothy NJ, Karlsson U, Reyes JF, Hallbeck M. Exposure of α-Synuclein Aggregates to Organotypic Slice Cultures Recapitulates Key Molecular Features of Parkinson's Disease. Front Neurol 2022; 13:826102. [PMID: 35309552 PMCID: PMC8925863 DOI: 10.3389/fneur.2022.826102] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/20/2022] [Indexed: 11/30/2022] Open
Abstract
The accumulation of proteinaceous deposits comprised largely of the α-synuclein protein is one of the main hallmarks of Parkinson's disease (PD) and related synucleinopathies. Their progressive development coincides with site-specific phosphorylation, oxidative stress and eventually, compromised neuronal function. However, modeling protein aggregate formation in animal or in vitro models has proven notably difficult. Here, we took advantage of a preclinical organotypic brain slice culture model to study α-synuclein aggregate formation ex vivo. We monitored the progressive and gradual changes induced by α-synuclein such as cellular toxicity, autophagy activation, mitochondrial dysfunction, cellular death as well as α-synuclein modification including site-specific phosphorylation. Our results demonstrate that organotypic brain slice cultures can be cultured for long periods of time and when cultured in the presence of aggregated α-synuclein, the molecular features of PD are recapitulated. Taken together, this ex vivo model allows for detailed modeling of the molecular features of PD, thus enabling studies on the cumulative effects of α-synuclein in a complex environment. This provides a platform to screen potential disease-modifying therapeutic candidates aimed at impeding α-synuclein aggregation and/or cellular transmission. Moreover, this model provides a robust replacement for in vivo studies that do not include behavioral experiments, thus providing a way to reduce the number of animals used in an accelerated timescale.
Collapse
Affiliation(s)
- Serge Moudio
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Rodin
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Nazira Jamal Albargothy
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Urban Karlsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Juan F Reyes
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Martin Hallbeck
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
13
|
Neonatal 6-hydroxydopamine lesioning of rats and dopaminergic neurotoxicity: proposed animal model of Parkinson’s disease. J Neural Transm (Vienna) 2022; 129:445-461. [DOI: 10.1007/s00702-022-02479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 10/18/2022]
|
14
|
Segura-Aguilar J, Mannervik B, Inzunza J, Varshney M, Nalvarte I, Muñoz P. Astrocytes protect dopaminergic neurons against aminochrome neurotoxicity. Neural Regen Res 2022; 17:1861-1866. [PMID: 35142659 PMCID: PMC8848618 DOI: 10.4103/1673-5374.335690] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology ICBM Faculty of Medicine University of Chile, Santiago, Chile
| | - Bengt Mannervik
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden
| | - José Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Patricia Muñoz
- Molecular and Clinical Pharmacology ICBM Faculty of Medicine University of Chile; Nucleo de Química y Bioquímica, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago, Chile
| |
Collapse
|
15
|
The Alpha-Synuclein RT-QuIC Products Generated by the Olfactory Mucosa of Patients with Parkinson’s Disease and Multiple System Atrophy Induce Inflammatory Responses in SH-SY5Y Cells. Cells 2021; 11:cells11010087. [PMID: 35011649 PMCID: PMC8750063 DOI: 10.3390/cells11010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/28/2022] Open
Abstract
Parkinson’s disease (PD) and multiple system atrophy (MSA) are caused by two distinct strains of disease-associated α-synuclein (αSynD). Recently, we have shown that olfactory mucosa (OM) samples of patients with PD and MSA can seed the aggregation of recombinant α-synuclein by means of Real-Time Quaking-Induced Conversion (αSyn_RT-QuIC). Remarkably, the biochemical and morphological properties of the final α-synuclein aggregates significantly differed between PD and MSA seeded samples. Here, these aggregates were given to neuron-like differentiated SH-SY5Y cells and distinct inflammatory responses were observed. To deepen whether the morphological features of α-synuclein aggregates were responsible for this variable SH-SY5Y inflammatory response, we generated three biochemically and morphologically distinct α-synuclein aggregates starting from recombinant α-synuclein that were used to seed αSyn_RT-QuIC reaction; the final reaction products were used to stimulate SH-SY5Y cells. Our study showed that, in contrast to OM samples of PD and MSA patients, the artificial aggregates did not transfer their distinctive features to the αSyn_RT-QuIC products and the latter induced analogous inflammatory responses in cells. Thus, the natural composition of the αSynD strains but also other specific factors in OM tissue can substantially modulate the biochemical, morphological and inflammatory features of the αSyn_RT-QuIC products.
Collapse
|
16
|
Yemula N, Dietrich C, Dostal V, Hornberger M. Parkinson's Disease and the Gut: Symptoms, Nutrition, and Microbiota. JOURNAL OF PARKINSON'S DISEASE 2021; 11:1491-1505. [PMID: 34250955 PMCID: PMC8609682 DOI: 10.3233/jpd-212707] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, characterized by symptoms of bradykinesia, rigidity, postural instability, and tremor. Recently, there has been a growing focus on the relationship between the gut and the development of PD. Emerging to the forefront, an interesting concept has developed suggesting that the initial pathophysiological changes occur in the gastrointestinal tract before changes are seen within the brain. This review is aimed at highlighting the relationship between PD and the gastrointestinal tract, along with the supporting evidence for this. Firstly, we will focus on the gastrointestinal conditions and symptoms which commonly affects patients, including both upper and lower gastrointestinal issues. Secondly, the impact of nutrition and diet on neurological health and PD physiology, with particular emphasis on commonly consumed items including macronutrients and micronutrients. Finally, variability of the gut microbiome will also be discussed and its link with both the symptoms and signs of PD. The evidence presented in this review highly suggests that the initial pathogenesis in the gut may proceed the development of prodromal PD subtypes, and therefore building on this further could be imperative and lead to earlier diagnosis with new and improved therapeutics.
Collapse
Affiliation(s)
- Nehal Yemula
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Celina Dietrich
- Faculty of Health and Medical Sciences, University of East Anglia, Norwich, United Kingdom
| | - Vaclav Dostal
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Michael Hornberger
- Faculty of Health and Medical Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
17
|
Mechanistic Insight from Preclinical Models of Parkinson's Disease Could Help Redirect Clinical Trial Efforts in GDNF Therapy. Int J Mol Sci 2021; 22:ijms222111702. [PMID: 34769132 PMCID: PMC8583859 DOI: 10.3390/ijms222111702] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by four pathognomonic hallmarks: (1) motor and non-motor deficits; (2) neuroinflammation and oxidative stress; (3) pathological aggregates of the α-synuclein (α-syn) protein; (4) neurodegeneration of the nigrostriatal system. Recent evidence sustains that the aggregation of pathological α-syn occurs in the early stages of the disease, becoming the first trigger of neuroinflammation and subsequent neurodegeneration. Thus, a therapeutic line aims at striking back α-synucleinopathy and neuroinflammation to impede neurodegeneration. Another therapeutic line is restoring the compromised dopaminergic system using neurotrophic factors, particularly the glial cell-derived neurotrophic factor (GDNF). Preclinical studies with GDNF have provided encouraging results but often lack evaluation of anti-α-syn and anti-inflammatory effects. In contrast, clinical trials have yielded imprecise results and have reported the emergence of severe side effects. Here, we analyze the discrepancy between preclinical and clinical outcomes, review the mechanisms of the aggregation of pathological α-syn, including neuroinflammation, and evaluate the neurorestorative properties of GDNF, emphasizing its anti-α-syn and anti-inflammatory effects in preclinical and clinical trials.
Collapse
|
18
|
Park SJ, Jin U, Park SM. Interaction between coxsackievirus B3 infection and α-synuclein in models of Parkinson's disease. PLoS Pathog 2021; 17:e1010018. [PMID: 34695168 PMCID: PMC8568191 DOI: 10.1371/journal.ppat.1010018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 11/04/2021] [Accepted: 10/08/2021] [Indexed: 01/04/2023] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases. PD is pathologically characterized by the death of midbrain dopaminergic neurons and the accumulation of intracellular protein inclusions called Lewy bodies or Lewy neurites. The major component of Lewy bodies is α-synuclein (α-syn). Prion-like propagation of α-syn has emerged as a novel mechanism in the progression of PD. This mechanism has been investigated to reveal factors that initiate Lewy pathology with the aim of preventing further progression of PD. Here, we demonstrate that coxsackievirus B3 (CVB3) infection can induce α-syn-associated inclusion body formation in neurons which might act as a trigger for PD. The inclusion bodies contained clustered organelles, including damaged mitochondria with α-syn fibrils. α-Syn overexpression accelerated inclusion body formation and induced more concentric inclusion bodies. In CVB3-infected mice brains, α-syn aggregates were observed in the cell body of midbrain neurons. Additionally, α-syn overexpression favored CVB3 replication and related cytotoxicity. α-Syn transgenic mice had a low survival rate, enhanced CVB3 replication, and exhibited neuronal cell death, including that of dopaminergic neurons in the substantia nigra. These results may be attributed to distinct autophagy-related pathways engaged by CVB3 and α-syn. This study elucidated the mechanism of Lewy body formation and the pathogenesis of PD associated with CVB3 infection. Prion-like propagation of α-syn has emerged as a novel mechanism involved in the progression of Parkinson’s disease (PD). This process has been extensively investigated to identify the factors that initiate Lewy pathology to prevent further progression of PD. Nevertheless, initial triggers of Lewy body (LB) formation leading to the acceleration of the process still remain elusive. Infection is increasingly recognized as a risk factor for PD. In particular, several viruses have been reported to be associated with both acute and chronic parkinsonism. It has been proposed that peripheral infections including viral infections accompanying inflammation may trigger PD. In the present study, we explored whether coxsackievirus B3 (CVB3) interacts with α-syn to induce aggregation and further Lewy body formation, thereby acting as a trigger and whether α-syn affects the replication of coxsackievirus. It is important to identify the factors that initiate Lewy pathology to understand the pathogenesis of PD. Our findings clarify the mechanism of LB formation and the pathogenesis of PD associated with CVB3 infection.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Uram Jin
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
- Department of Cardiology, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
- * E-mail:
| |
Collapse
|
19
|
Sevgi F, Brauchle EM, Carvajal Berrio DA, Schenke-Layland K, Casadei N, Salker MS, Riess O, Singh Y. Imaging of α-Synuclein Aggregates in a Rat Model of Parkinson's Disease Using Raman Microspectroscopy. Front Cell Dev Biol 2021; 9:664365. [PMID: 34568310 PMCID: PMC8461246 DOI: 10.3389/fcell.2021.664365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/14/2021] [Indexed: 11/18/2022] Open
Abstract
A hallmark of Parkinson’s disease (PD) is the formation of Lewy bodies in the brain. Lewy bodies are rich in the aggregated form of misfolded α-Synuclein (α-Syn). The brain from PD patients can only be analyzed after postmortem, therefore, limiting the diagnosis of PD to the manifestation of motor symptoms. In PD patients and animal models, phosphorylated α-Syn was detected in the peripheral tissues including the gut, thus, raising the hypothesis that early-stage PD could be diagnosed based on colon tissue biopsies. Non-invasive marker-free technologies represent ideal methods to potentially detect aggregated α-Syn in vivo. Raman microspectroscopy has been established for the detection of molecular changes such as alterations of protein structures. Using Raman imaging and microspectroscopy, we analyzed the olfactory bulb in the brain and the muscularis mucosae of colon tissue sections of a human BAC-SNCA transgenic (TG) rat model. Raman images from TG and WT rats were investigated using principal component analysis (PCA) and true component analysis (TCA). Spectral components indicated protein aggregates (spheroidal oligomers) in the TG rat brain and in the colon tissues even at a young age but not in WT. In summary, we have demonstrated that Raman imaging is capable of detecting α-Syn aggregates in colon tissues of a PD rat model and making it a promising tool for future use in PD pathology.
Collapse
Affiliation(s)
- Fide Sevgi
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany
| | - Eva M Brauchle
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany.,Natural and Medical Sciences Institute (NMI), Tübingen University, Reutlingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel A Carvajal Berrio
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany.,Natural and Medical Sciences Institute (NMI), Tübingen University, Reutlingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls Tübingen University, Tübingen, Germany
| | - Madhuri S Salker
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls Tübingen University, Tübingen, Germany
| | - Yogesh Singh
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls Tübingen University, Tübingen, Germany.,Institute of Medical Genetics and Applied Genomics, Eberhard Karls Tübingen University, Tübingen, Germany
| |
Collapse
|
20
|
Sarchione A, Marchand A, Taymans JM, Chartier-Harlin MC. Alpha-Synuclein and Lipids: The Elephant in the Room? Cells 2021; 10:2452. [PMID: 34572099 PMCID: PMC8467310 DOI: 10.3390/cells10092452] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/17/2022] Open
Abstract
Since the initial identification of alpha-synuclein (α-syn) at the synapse, numerous studies demonstrated that α-syn is a key player in the etiology of Parkinson's disease (PD) and other synucleinopathies. Recent advances underline interactions between α-syn and lipids that also participate in α-syn misfolding and aggregation. In addition, increasing evidence demonstrates that α-syn plays a major role in different steps of synaptic exocytosis. Thus, we reviewed literature showing (1) the interplay among α-syn, lipids, and lipid membranes; (2) advances of α-syn synaptic functions in exocytosis. These data underscore a fundamental role of α-syn/lipid interplay that also contributes to synaptic defects in PD. The importance of lipids in PD is further highlighted by data showing the impact of α-syn on lipid metabolism, modulation of α-syn levels by lipids, as well as the identification of genetic determinants involved in lipid homeostasis associated with α-syn pathologies. While questions still remain, these recent developments open the way to new therapeutic strategies for PD and related disorders including some based on modulating synaptic functions.
Collapse
Affiliation(s)
| | | | | | - Marie-Christine Chartier-Harlin
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172—LilNCog—Lille Neuroscience and Cognition, F-59000 Lille, France; (A.S.); (A.M.); (J.-M.T.)
| |
Collapse
|
21
|
Xu L, Nagai Y, Kajihara Y, Ito G, Tomita T. The Regulation of Rab GTPases by Phosphorylation. Biomolecules 2021; 11:biom11091340. [PMID: 34572553 PMCID: PMC8469595 DOI: 10.3390/biom11091340] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 01/11/2023] Open
Abstract
Rab proteins are small GTPases that act as molecular switches for intracellular vesicle trafficking. Although their function is mainly regulated by regulatory proteins such as GTPase-activating proteins and guanine nucleotide exchange factors, recent studies have shown that some Rab proteins are physiologically phosphorylated in the switch II region by Rab kinases. As the switch II region of Rab proteins undergoes a conformational change depending on the bound nucleotide, it plays an essential role in their function as a ‘switch’. Initially, the phosphorylation of Rab proteins in the switch II region was shown to inhibit the association with regulatory proteins. However, recent studies suggest that it also regulates the binding of Rab proteins to effector proteins, determining which pathways to regulate. These findings suggest that the regulation of the Rab function may be more dynamically regulated by phosphorylation than just through the association with regulatory proteins. In this review, we summarize the recent findings and discuss the physiological and pathological roles of Rab phosphorylation.
Collapse
Affiliation(s)
- Lejia Xu
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Yuki Nagai
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Yotaro Kajihara
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Genta Ito
- Department of Biomolecular Chemistry, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Correspondence: (G.I.); (T.T.)
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Correspondence: (G.I.); (T.T.)
| |
Collapse
|
22
|
Ha TY, Choi YR, Noh HR, Cha SH, Kim JB, Park SM. Age-related increase in caveolin-1 expression facilitates cell-to-cell transmission of α-synuclein in neurons. Mol Brain 2021; 14:122. [PMID: 34321069 PMCID: PMC8320051 DOI: 10.1186/s13041-021-00834-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.
Collapse
Affiliation(s)
- Tae-Young Ha
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Hye Rin Noh
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Seon-Heui Cha
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Marine Biomedical Sciences, Hanseo University, Seosan, Chungcheongnam-do, Korea
| | - Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
23
|
Beekes M. The Neural Gut-Brain Axis of Pathological Protein Aggregation in Parkinson's Disease and Its Counterpart in Peroral Prion Infections. Viruses 2021; 13:1394. [PMID: 34372600 PMCID: PMC8310171 DOI: 10.3390/v13071394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
A neuropathological hallmark of Parkinson's disease (PD) is the cerebral deposition of abnormally aggregated α-synuclein (αSyn). PD-associated αSyn (αSynPD) aggregates are assumed to act, in a prion-like manner, as proteinaceous nuclei ("seeds") capable of self-templated propagation. Braak and colleagues put forward the idea of a neural gut-brain axis mediating the centripetal spread of αSynPD pathology from the enteric nervous system (ENS) to the brain in PD. This has sparked great interest and initiated passionate discussions both in support of and opposing the suggested hypothesis. A precedent for the spread of protein seeds or seeding from the gastro-intestinal (GI) tract to the central nervous system (CNS) had been previously revealed for pathological prion protein in peroral prion infections. This article scrutinizes the similarities and dissimilarities between the pathophysiological spread of disease-associated protein aggregation along the neural gut-brain axis in peroral prion infections and PD. On this basis, evidence supporting the proposed neural gut-brain axis in PD is concluded to be not as robust as that established for peroral prion infections. New tools for the ultrasensitive detection of αSynPD-associated seeding activity in archived or fresh human tissue samples such as real-time quaking induced conversion (RT-QuIC) or protein misfolding cyclic amplification (PMCA) assays can possibly help to address this deficit in the future.
Collapse
Affiliation(s)
- Michael Beekes
- Prion and Prionoid Research Unit, ZBS 6-Proteomics and Spectroscopy, ZBS-Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| |
Collapse
|
24
|
García‐Sanz P, M.F.G. Aerts J, Moratalla R. The Role of Cholesterol in α-Synuclein and Lewy Body Pathology in GBA1 Parkinson's Disease. Mov Disord 2021; 36:1070-1085. [PMID: 33219714 PMCID: PMC8247417 DOI: 10.1002/mds.28396] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease where dopaminergic neurons in the substantia nigra are lost, resulting in a decrease in striatal dopamine and, consequently, motor control. Dopaminergic degeneration is associated with the appearance of Lewy bodies, which contain membrane structures and proteins, including α-synuclein (α-Syn), in surviving neurons. PD displays a multifactorial pathology and develops from interactions between multiple elements, such as age, environmental conditions, and genetics. Mutations in the GBA1 gene represent one of the major genetic risk factors for PD. This gene encodes an essential lysosomal enzyme called β-glucocerebrosidase (GCase), which is responsible for degrading the glycolipid glucocerebroside into glucose and ceramide. GCase can generate glucosylated cholesterol via transglucosylation and can also degrade the sterol glucoside. Although the molecular mechanisms that predispose an individual to neurodegeneration remain unknown, the role of cholesterol in PD pathology deserves consideration. Disturbed cellular cholesterol metabolism, as reflected by accumulation of lysosomal cholesterol in GBA1-associated PD cellular models, could contribute to changes in lipid rafts, which are necessary for synaptic localization and vesicle cycling and modulation of synaptic integrity. α-Syn has been implicated in the regulation of neuronal cholesterol, and cholesterol facilitates interactions between α-Syn oligomers. In this review, we integrate the results of previous studies and describe the cholesterol landscape in cellular homeostasis and neuronal function. We discuss its implication in α-Syn and Lewy body pathophysiological mechanisms underlying PD, focusing on the role of GCase and cholesterol. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patricia García‐Sanz
- Instituto Cajal, CSICMadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| | - Johannes M.F.G. Aerts
- Medical Biochemistry, Leiden Institute of Chemistry, Leiden UniversityFaculty of ScienceLeidenthe Netherlands
| | - Rosario Moratalla
- Instituto Cajal, CSICMadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasInstituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
25
|
Zhu C, Bilousova T, Focht S, Jun M, Elias CJ, Melnik M, Chandra S, Campagna J, Cohn W, Hatami A, Spilman P, Gylys KH, John V. Pharmacological inhibition of nSMase2 reduces brain exosome release and α-synuclein pathology in a Parkinson's disease model. Mol Brain 2021; 14:70. [PMID: 33875010 PMCID: PMC8056538 DOI: 10.1186/s13041-021-00776-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Aim We have previously reported that cambinol (DDL-112), a known inhibitor of neutral sphingomyelinase-2 (nSMase2), suppressed extracellular vesicle (EV)/exosome production in vitro in a cell model and reduced tau seed propagation. The enzyme nSMase2 is involved in the production of exosomes carrying proteopathic seeds and could contribute to cell-to-cell transmission of pathological protein aggregates implicated in neurodegenerative diseases such as Parkinson’s disease (PD). Here, we performed in vivo studies to determine if DDL-112 can reduce brain EV/exosome production and proteopathic alpha synuclein (αSyn) spread in a PD mouse model. Methods The acute effects of single-dose treatment with DDL-112 on interleukin-1β-induced extracellular vesicle (EV) release in brain tissue of Thy1-αSyn PD model mice and chronic effects of 5 week DDL-112 treatment on behavioral/motor function and proteinase K-resistant αSyn aggregates in the PD model were determined. Results/discussion In the acute study, pre-treatment with DDL-112 reduced EV/exosome biogenesis and in the chronic study, treatment with DDL-112 was associated with a reduction in αSyn aggregates in the substantia nigra and improvement in motor function. Inhibition of nSMase2 thus offers a new approach to therapeutic development for neurodegenerative diseases with the potential to reduce the spread of disease-specific proteopathic proteins. Supplementary Information The online version contains supplementary material available at 10.1186/s13041-021-00776-9.
Collapse
Affiliation(s)
- Chunni Zhu
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Tina Bilousova
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA.,School of Nursing, University of California, Los Angeles, CA, 90095, USA
| | - Samantha Focht
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Michael Jun
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Chris Jean Elias
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Mikhail Melnik
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Sujyoti Chandra
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Jesus Campagna
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Whitaker Cohn
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Asa Hatami
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | - Patricia Spilman
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA
| | | | - Varghese John
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
26
|
Dong TTT, Satoh K. The Latest Research on RT-QuIC Assays-A Literature Review. Pathogens 2021; 10:pathogens10030305. [PMID: 33807776 PMCID: PMC8000803 DOI: 10.3390/pathogens10030305] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
The misfolding of proteins such as the prion protein, α-synuclein, and tau represents a key initiating event for pathogenesis of most common neurodegenerative disorders, and its presence correlates with infectivity. To date, the diagnosis of these disorders mainly relied on the recognition of clinical symptoms when neurodegeneration was already at an advanced phase. In recent years, several efforts have been made to develop new diagnostic tools for the early diagnosis of prion diseases. The real-time quaking-induced conversion (RT–QuIC) assay, an in vitro assay that can indirectly detect very low amounts of PrPSc aggregates, has provided a very promising tool to improve the early diagnosis of human prion diseases. Over the decade since RT–QuIC was introduced, the diagnosis of not only prion diseases but also synucleinopathies and tauopathies has greatly improved. Therefore, in our study, we summarize the current trends and knowledge of RT–QuIC assays, as well as discuss the diagnosis of neurodegenerative diseases using RT–QuIC assays, which have been updated in recent years.
Collapse
|
27
|
Asher DM, Belay E, Bigio E, Brandner S, Brubaker SA, Caughey B, Clark B, Damon I, Diamond M, Freund M, Hyman BT, Jucker M, Keene CD, Lieberman AP, Mackiewicz M, Montine TJ, Morgello S, Phelps C, Safar J, Schneider JA, Schonberger LB, Sigurdson C, Silverberg N, Trojanowski JQ, Frosch MP. Risk of Transmissibility From Neurodegenerative Disease-Associated Proteins: Experimental Knowns and Unknowns. J Neuropathol Exp Neurol 2021; 79:1141-1146. [PMID: 33000167 PMCID: PMC7577514 DOI: 10.1093/jnen/nlaa109] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent studies in animal models demonstrate that certain misfolded proteins associated with neurodegenerative diseases can support templated misfolding of cognate native proteins, to propagate across neural systems, and to therefore have some of the properties of classical prion diseases like Creutzfeldt-Jakob disease. The National Institute of Aging convened a meeting to discuss the implications of these observations for research priorities. A summary of the discussion is presented here, with a focus on limitations of current knowledge, highlighting areas that appear to require further investigation in order to guide scientific practice while minimizing potential exposure or risk in the laboratory setting. The committee concluded that, based on all currently available data, although neurodegenerative disease-associated aggregates of several different non-prion proteins can be propagated from humans to experimental animals, there is currently insufficient evidence to suggest more than a negligible risk, if any, of a direct infectious etiology for the human neurodegenerative disorders defined in part by these proteins. Given the importance of this question, the potential for noninvasive human transmission of proteopathic disorders is deserving of further investigation.
Collapse
Affiliation(s)
- David M Asher
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Ermias Belay
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Eileen Bigio
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology Queen Square, London
| | - Scott A Brubaker
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Brychan Clark
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Inger Damon
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Marc Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michelle Freund
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen and German Center for Neurodegenerative Diseases (DZNE), Tübingen
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Miroslaw Mackiewicz
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California
| | - Susan Morgello
- Departments of Neurology, Neuroscience, and Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Creighton Phelps
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Jiri Safar
- Departments of Pathology and Neurology, Case Western Reserve University, Cleveland, Ohio
| | - Julie A Schneider
- Department of Neurological Sciences, Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Lawrence B Schonberger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christina Sigurdson
- Department of Pathology, University of California - San Diego, San Diego, California
| | - Nina Silverberg
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthew P Frosch
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, University of Washington, Seattle, Washington.,C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
28
|
Derkinderen P, Rolli-Derkinderen M, Chapelet G, Neunlist M, Noble W. Tau in the gut, does it really matter? J Neurochem 2021; 158:94-104. [PMID: 33569813 DOI: 10.1111/jnc.15320] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022]
Abstract
The enteric nervous system plays a critical role in the regulation of gastrointestinal tract functions and is often referred to as the 'second brain' because it shares many features with the central nervous system. These similarities include among others a large panel of neurotransmitters, a large population of glial cells and a susceptibility to neurodegeneration. This close homology between the central and enteric nervous systems suggests that a disease process affecting the central nervous system could also involve its enteric counterpart. This was already documented in Parkinson's disease, the most common synucleinopathy, in which alpha-synuclein deposits are reported in the enteric nervous system in the vast majority of patients. Tau is another key protein involved in neurodegenerative disorders of the brain. Whether changes in tau also occur in the enteric nervous system during gut or brain disorders has just begun to be explored. The scope of the present article is therefore to review existing studies on the expression and phosphorylation pattern of tau in the enteric nervous system under physiological and pathological conditions and to discuss the possible occurrence of 'enteric tauopathies'.
Collapse
Affiliation(s)
- Pascal Derkinderen
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Malvyne Rolli-Derkinderen
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Guillaume Chapelet
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France.,Clinical Gerontology Department, CHU Nantes, Nantes, France
| | - Michel Neunlist
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| |
Collapse
|
29
|
Wang T, Shi C, Luo H, Zheng H, Fan L, Tang M, Su Y, Yang J, Mao C, Xu Y. Neuroinflammation in Parkinson's Disease: Triggers, Mechanisms, and Immunotherapies. Neuroscientist 2021; 28:364-381. [PMID: 33576313 DOI: 10.1177/1073858421991066] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease involving multiple etiologies and pathogenesis, in which neuroinflammation is a common factor. Both preclinical experiments and clinical studies provide evidence for the involvement of neuroinflammation in the pathophysiology of PD, although there are a number of key issues related to neuroinflammatory processes in PD that remain to be addressed. In this review, we highlight the relationship between the common pathological mechanisms of PD and neuroinflammation, including aggregation of α-synuclein, genetic factors, mitochondrial dysfunction, and gut microbiome dysbiosis. We also describe the two positive feedback loops initiated in PD after the immune system is activated, and their role in the pathogenesis of PD. In addition, the interconnections and differences between the central and peripheral immune systems are discussed. Finally, we review the latest progress in immunotherapy research for PD patients, and propose future directions for clinical research.
Collapse
Affiliation(s)
- Tai Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
30
|
Wu M, Su H, Zhao M. The Role of α-Synuclein in Methamphetamine-Induced Neurotoxicity. Neurotox Res 2021; 39:1007-1021. [PMID: 33555547 DOI: 10.1007/s12640-021-00332-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/28/2022]
Abstract
Methamphetamine (METH), a highly addictive psychostimulant, is the second most widely used illicit drug. METH produces damage dopamine neurons and apoptosis via multiple inter-regulating mechanisms, including dopamine overload, hyperthermia, oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, protein degradation system dysfunction, and neuroinflammation. Increasing evidence suggests that chronic METH abuse is associated with neurodegenerative changes in the human brain and an increased risk of Parkinson's disease (PD). METH use and PD may share some common steps in causing neurotoxicity. Accumulation of α-synuclein, a presynaptic protein, is the pathological hallmark of PD. Intriguingly, α-synuclein upregulation and aggregation are also found in dopaminergic neurons in the substantia nigra in chronic METH users. This suggests α-synuclein may play a role in METH-induced neurotoxicity. The mechanism of α-synuclein cytotoxicity in PD has attracted considerable attention; however, how α-synuclein affects METH-induced neurotoxicity has not been reviewed. In this review, we summarize the relationship between METH use and PD, interdependent mechanisms that are involved in METH-induced neurotoxicity and the significance of α-synuclein upregulation in response to METH use. The identification of α-synuclein overexpression and aggregation as a contributor to METH-induced neurotoxicity may provide a novel therapeutic target for the treatment of the deleterious effect of this drug and drug addiction.
Collapse
Affiliation(s)
- Manqing Wu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang Su
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- Shanghai Clinical Research Center for Mental Health, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
31
|
Timmerman R, Burm SM, Bajramovic JJ. Tissue-specific features of microglial innate immune responses. Neurochem Int 2020; 142:104924. [PMID: 33248205 DOI: 10.1016/j.neuint.2020.104924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
As tissue-resident macrophages of the brain, microglia are increasingly considered as cellular targets for therapeutical intervention. Innate immune responses in particular have been implicated in central nervous system (CNS) infections, neuro-oncology, neuroinflammatory and neurodegenerative diseases. We here review the impact of 'nature and nurture' on microglial innate immune responses and summarize documented tissue-specific adaptations. Overall, such adaptations are associated with regulatory processes rather than with overt differences in the expressed repertoire of activating receptors of different tissue-resident macrophages. Microglial responses are characterized by slower kinetics, by a more persistent nature and by a differential usage of downstream enzymes and accessory receptors. We further consider factors like aging, previous exposure to inflammatory stimuli, and differences in the microenvironment that can modulate innate immune responses. The long-life span of microglia in the metabolically active CNS renders them susceptible to the phenomenon of 'inflammaging', and major challenges lie in the unraveling of the factors that underlie age-related alterations in microglial behavior.
Collapse
Affiliation(s)
- R Timmerman
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - S M Burm
- Genmab, Utrecht, the Netherlands
| | - J J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| |
Collapse
|
32
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Patnaik R, Wiklund L, Sharma HS. Co-administration of TiO 2-nanowired dl-3-n-butylphthalide (dl-NBP) and mesenchymal stem cells enhanced neuroprotection in Parkinson's disease exacerbated by concussive head injury. PROGRESS IN BRAIN RESEARCH 2020; 258:101-155. [PMID: 33223034 DOI: 10.1016/bs.pbr.2020.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
dl-3-n-butylphthalide (dl-NBP) is a powerful antioxidant compound with profound neuroprotective effects in stroke and brain injury. However, its role in Parkinson's disease (PD) is not well known. Traumatic brain injury (TBI) is one of the key factors in precipitating PD like symptoms in civilians and particularly in military personnel. Thus, it would be interesting to explore the possible neuroprotective effects of NBP in PD following concussive head injury (CHI). In this chapter effect of nanowired delivery of NBP together with mesenchymal stem cells (MSCs) in PD with CHI is discussed based on our own investigations. It appears that CHI exacerbates PD pathophysiology in terms of p-tau, α-synuclein (ASNC) levels in the cerebrospinal fluid (CSF) and the loss of TH immunoreactivity in substantia niagra pars compacta (SNpc) and striatum (STr) along with dopamine (DA), dopamine decarboxylase (DOPAC). And homovanillic acid (HVA). Our observations are the first to show that a combination of NBP with MSCs when delivered using nanowired technology induces superior neuroprotective effects in PD brain pathology exacerbated by CHI, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, Hebei Province, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, Hebei Province, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
33
|
Remodeling microglia to a protective phenotype in Parkinson's disease? Neurosci Lett 2020; 735:135164. [PMID: 32561452 DOI: 10.1016/j.neulet.2020.135164] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the most widespread movement disorder with a prevalence of 1 in 1000 individuals above 60 years of age. Until now, understanding the pathological mechanisms of PD to translate them into therapy has remained a high research priority. In this review, we highlight evidence describing the involvement of microglial dysfunction in PD. Thereafter, we provide current knowledge suggesting that the substantia nigra pars compacta and putamen, compared to other brain regions, show a reduced microglial density, as well as altered morphological and functional properties in homeostatic conditions, while presenting dystrophic features associated with aging. Further, we describe that this defective microglial programing emerges as early as the second postnatal week, persists until adulthood and impacts negatively on their transcriptional pattern and provision of local trophic support. We emphasize the role of α-synuclein oligomers as a major dysfunctional signal underlining microglial-mediated phenotypic switch and adaptive response contributing to neurodegeneration. Moreover, we explore available avenues should microglia be considered as target for neuroprotective or restorative strategies including preventing the aggregation of α-synuclein protofibrils formation. However, we provide a note of caution regarding the success of microglial-targeted PD strategies, using minocycline as an example. In conclusion, we discuss putative neuroprotective agents that were unsuccessful in previous trials but could be reconsidered by focusing on the stage of microglial-dependent pathogenic events during PD in suitable cohorts of patients.
Collapse
|
34
|
Johnson ME, Bergkvist L, Mercado G, Stetzik L, Meyerdirk L, Wolfrum E, Madaj Z, Brundin P, Wesson DW. Deficits in olfactory sensitivity in a mouse model of Parkinson's disease revealed by plethysmography of odor-evoked sniffing. Sci Rep 2020; 10:9242. [PMID: 32514004 PMCID: PMC7280205 DOI: 10.1038/s41598-020-66201-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/18/2020] [Indexed: 12/26/2022] Open
Abstract
Hyposmia is evident in over 90% of Parkinson’s disease (PD) patients. A characteristic of PD is intraneuronal deposits composed in part of α-synuclein fibrils. Based on the analysis of post-mortem PD patients, Braak and colleagues suggested that early in the disease α-synuclein pathology is present in the dorsal motor nucleus of the vagus, as well as the olfactory bulb and anterior olfactory nucleus, and then later affects other interconnected brain regions. Here, we bilaterally injected α-synuclein preformed fibrils into the olfactory bulbs of wild type male and female mice. Six months after injection, the anterior olfactory nucleus and piriform cortex displayed a high α-synuclein pathology load. We evaluated olfactory perceptual function by monitoring odor-evoked sniffing behavior in a plethysmograph at one-, three- and six-months after injection. No overt impairments in the ability to engage in sniffing were evident in any group, suggesting preservation of the ability to coordinate respiration. At all-time points, females injected with fibrils exhibited reduced odor detection sensitivity, which was observed with the semi-automated plethysmography apparatus, but not a buried pellet test. In future studies, this sensitive methodology for assessing olfactory detection deficits could be used to define how α-synuclein pathology affects other aspects of olfactory perception and to clarify the neuropathological underpinnings of these deficits.
Collapse
Affiliation(s)
- Michaela E Johnson
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Liza Bergkvist
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Gabriela Mercado
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Lucas Stetzik
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Lindsay Meyerdirk
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Emily Wolfrum
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503, US
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, US.
| | - Daniel W Wesson
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
35
|
Choi YR, Kim JB, Kang SJ, Noh HR, Jou I, Joe EH, Park SM. The dual role of c-src in cell-to-cell transmission of α-synuclein. EMBO Rep 2020; 21:e48950. [PMID: 32372484 DOI: 10.15252/embr.201948950] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/02/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons located in the substantia nigra pars compacta and the presence of proteinaceous inclusions called Lewy bodies and Lewy neurites in numerous brain regions. Increasing evidence indicates that Lewy pathology progressively involves additional regions of the nervous system as the disease advances, and the prion-like propagation of α-synuclein (α-syn) pathology promotes PD progression. Accordingly, the modulation of α-syn transmission may be important for the development of disease-modifying therapies in patients with PD. Here, we demonstrate that α-syn fibrils induce c-src activation in neurons, which depends on the FcγRIIb-SHP-1/-2-c-src pathway and enhances signals for the uptake of α-syn into neurons. Blockade of c-src activation inhibits the uptake of α-syn and the formation of Lewy body-like inclusions. Furthermore, the blockade of c-src activation also inhibits the release of α-syn via activation of autophagy. The brain-permeable c-src inhibitor, saracatinib, efficiently reduces α-syn propagation into neighboring regions in an in vivo model system. These results suggest a new therapeutic target against progressive PD.
Collapse
Affiliation(s)
- Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Seo-Jun Kang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Hye Rin Noh
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Ilo Jou
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, BK21 Plus Program, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
36
|
Hirsch EC, Standaert DG. Ten Unsolved Questions About Neuroinflammation in Parkinson's Disease. Mov Disord 2020; 36:16-24. [PMID: 32357266 DOI: 10.1002/mds.28075] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease is a progressive and debilitating disorder that has so far eluded attempts to develop disease-modifying treatment. Both epidemiological and genetic studies support a role of neuroinflammation in the pathophysiology of Parkinson's disease. Postmortem studies and experimental analyses suggest the involvement of both innate and adaptive immunity in the degenerative process. There is also some circumstantial evidence for effects of immune therapies on the disease. In the present article, we review 10 unanswered questions related to neuroinflammatory processes in Parkinson's disease with the goal of stimulating research in the field and accelerating the clinical development of neuroprotective therapies based on anti-inflammatory strategies. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Etienne C Hirsch
- Faculté de Médecine de Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, U 1127, CNRS, Unité Mixte de Recherche (UMR) 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
37
|
Soto-Rojas LO, Martínez-Dávila IA, Luna-Herrera C, Gutierrez-Castillo ME, Lopez-Salas FE, Gatica-Garcia B, Soto-Rodriguez G, Bringas Tobon ME, Flores G, Padilla-Viveros A, Bañuelos C, Blanco-Alvarez VM, Dávila-Ayala J, Reyes-Corona D, Garcés-Ramírez L, Hidalgo-Alegria O, De La Cruz-lópez F, Martinez-Fong D. Unilateral intranigral administration of β-sitosterol β-D-glucoside triggers pathological α-synuclein spreading and bilateral nigrostriatal dopaminergic neurodegeneration in the rat. Acta Neuropathol Commun 2020; 8:56. [PMID: 32321590 PMCID: PMC7178762 DOI: 10.1186/s40478-020-00933-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/14/2020] [Indexed: 02/05/2023] Open
Abstract
The spreading and accumulation of α-synuclein and dopaminergic neurodegeneration, two hallmarks of Parkinson’s disease (PD), have been faithfully reproduced in rodent brains by chronic, oral administration of β-sitosterol β-D-glucoside (BSSG). We investigated whether a single injection of BSSG (6 μg BSSG/μL DMSO) in the left substantia nigra of Wistar rats causes the same effects. Mock DMSO injections and untreated rats formed control groups. We performed immunostainings against the pathological α-synuclein, the dopaminergic marker tyrosine hydroxylase (TH), the neuroskeleton marker β-III tubulin, the neurotensin receptor type 1 (NTSR1) as non-dopaminergic phenotype marker and Fluro-Jade C (F-J C) label for neurodegeneration. Using β-galactosidase (β-Gal) assay and active caspase-3 immunostaining, we assessed cell death mechanisms. Golgi-Cox staining was used to measure the density and types of dendritic spines of striatal medium spiny neurons. Motor and non-motor alterations were also evaluated. The study period comprised 15 to 120 days after the lesion. In the injured substantia nigra, BSSG caused a progressive α-synuclein aggregation and dopaminergic neurodegeneration caused by senescence and apoptosis. The α-synuclein immunoreactivity was also present within microglia cells. Decreased density of dopaminergic fibers and dendritic spines also occurred in the striatum. Remarkably, all the histopathological changes also appeared on the contralateral nigrostriatal system, and α-synuclein aggregates were present in other brain regions. Motor and non-motor behavioral alterations were progressive. Our data show that the stereotaxic BSSG administration reproduces PD α-synucleinopathy phenotype in the rat. This approach will aid in identifying the spread mechanism of α-synuclein pathology and validate anti-synucleinopathy therapies.
Collapse
|
38
|
Rozpędek-Kamińska W, Siwecka N, Wawrzynkiewicz A, Wojtczak R, Pytel D, Diehl JA, Majsterek I. The PERK-Dependent Molecular Mechanisms as a Novel Therapeutic Target for Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E2108. [PMID: 32204380 PMCID: PMC7139310 DOI: 10.3390/ijms21062108] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
Higher prevalence of neurodegenerative diseases is strictly connected with progressive aging of the world population. Interestingly, a broad range of age-related, neurodegenerative diseases is characterized by a common pathological mechanism-accumulation of misfolded and unfolded proteins within the cells. Under certain circumstances, such protein aggregates may evoke endoplasmic reticulum (ER) stress conditions and subsequent activation of the unfolded protein response (UPR) signaling pathways via the protein kinase RNA-like endoplasmic reticulum kinase (PERK)-dependent manner. Under mild to moderate ER stress, UPR has a pro-adaptive role. However, severe or long-termed ER stress conditions directly evoke shift of the UPR toward its pro-apoptotic branch, which is considered to be a possible cause of neurodegeneration. To this day, there is no effective cure for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), or prion disease. Currently available treatment approaches for these diseases are only symptomatic and cannot affect the disease progression. Treatment strategies, currently under detailed research, include inhibition of the PERK-dependent UPR signaling branches. The newest data have reported that the use of small-molecule inhibitors of the PERK-mediated signaling branches may contribute to the development of a novel, ground-breaking therapeutic approach for neurodegeneration. In this review, we critically describe all the aspects associated with such targeted therapy against neurodegenerative proteopathies.
Collapse
Affiliation(s)
- Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Adam Wawrzynkiewicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Radosław Wojtczak
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Dariusz Pytel
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; (D.P.); (J.A.D.)
| | - J. Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; (D.P.); (J.A.D.)
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| |
Collapse
|
39
|
Stott SRW, Wyse RK, Brundin P. Novel approaches to counter protein aggregation pathology in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:451-492. [PMID: 32247372 PMCID: PMC10019778 DOI: 10.1016/bs.pbr.2019.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The primary neuropathological characteristics of the Parkinsonian brain are the loss of nigral dopamine neurons and the aggregation of alpha synuclein protein. Efforts to development potentially disease-modifying treatments have largely focused on correcting these aspects of the condition. In the last decade treatments targeting protein aggregation have entered the clinical pipeline. In this chapter we provide an overview of ongoing clinical trial programs for different therapies attempting to reduce protein aggregation pathology in Parkinson's disease. We will also briefly consider various novel approaches being proposed-and being developed preclinically-to inhibit/reduce aggregated protein pathology in Parkinson's.
Collapse
Affiliation(s)
| | | | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
40
|
Fields CR, Bengoa-Vergniory N, Wade-Martins R. Targeting Alpha-Synuclein as a Therapy for Parkinson's Disease. Front Mol Neurosci 2019; 12:299. [PMID: 31866823 PMCID: PMC6906193 DOI: 10.3389/fnmol.2019.00299] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 01/23/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders with a global burden of approximately 6.1 million patients. Alpha-synuclein has been linked to both the sporadic and familial forms of the disease. Moreover, alpha-synuclein is present in Lewy-bodies, the neuropathological hallmark of PD, and the protein and its aggregation have been widely linked to neurotoxic pathways that ultimately lead to neurodegeneration. Such pathways include autophagy/lysosomal dysregulation, synaptic dysfunction, mitochondrial disruption, and endoplasmic reticulum (ER) and oxidative stress. Alpha-synuclein has not only been shown to alter cellular pathways but also to spread between cells, causing aggregation in host cells. Therapeutic approaches will need to address several, if not all, of these angles of alpha-synuclein toxicity. Here we review the current advances in therapeutic efforts for PD that aim to produce a disease-modifying therapy by targeting the spread, production, aggregation, and degradation of alpha-synuclein. These include: receptor blocking strategies whereby putative alpha-synuclein receptors could be blocked inhibiting alpha-synuclein spread, an alpha-synuclein reduction which will decrease the amount alpha-synuclein available for aggregation and pathway disruption, the use of small molecules in order to target alpha-synuclein aggregation, immunotherapy and the increase of alpha-synuclein degradation by increasing autophagy/lysosomal flux. The research discussed here may lead to a disease-modifying therapy that tackles disease onset and progression in the future.
Collapse
Affiliation(s)
| | - Nora Bengoa-Vergniory
- Department of Physiology, Oxford Parkinson's Disease Center, Anatomy and Genetics, Oxford, United Kingdom
| | - Richard Wade-Martins
- Department of Physiology, Oxford Parkinson's Disease Center, Anatomy and Genetics, Oxford, United Kingdom
| |
Collapse
|
41
|
Longhena F, Faustini G, Brembati V, Pizzi M, Bellucci A. The good and bad of therapeutic strategies that directly target α-synuclein. IUBMB Life 2019; 72:590-600. [PMID: 31693290 DOI: 10.1002/iub.2194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/12/2019] [Indexed: 12/16/2022]
Abstract
Synucleinopathies are neurodegenerative diseases characterized by the accumulation of either neuronal/axonal or glial insoluble proteinaceous aggregates mainly composed of α-synuclein (α-syn). Among them, the most common disorders are Parkinson's disease, dementia with Lewy bodies, multiple system atrophy, and some forms of familial parkinsonism. Both α-syn fibrils and oligomers have been found to exert toxic effects on neurons or oligodendroglial cells, can activate neuroinflammatory responses, and mediate the spreading of α-syn pathology. This poses the question of which is the most toxic α-syn species. What is worst, α-syn appears as a very peculiar protein, exerting multiple physiological functions in neurons, especially at synapses, but without acquiring a stable tertiary structure. Its conformation is particularly plastic, and the protein can exist in a natively unfolded state (mainly in solution), partially α-helical folded state (when it interacts with biological membranes), or oligomeric state (tetramers or dimers with debated functional profile). The extent of α-syn expression impinges on the resilience of neuronal cells, as multiplications of its gene locus, or overexpression, can cause neurodegeneration and onset of motor phenotype. For these reasons, one of the main challenges in the field of synucleinopathies, which still nowadays can only be managed by symptomatic therapies, has been the development of strategies aimed at reducing α-syn levels, oligomer formation, fibrillation, or cell-to-cell transmission. This review resumes the therapeutic approaches that have been proposed or are under development to counteract α-syn pathology by direct targeting of this protein and discuss their pros and cons in relation to the current state-of-the-art α-syn biology.
Collapse
Affiliation(s)
- Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Viviana Brembati
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
42
|
Wong Y, Luk K, Purtell K, Nanni SB, Stoessl AJ, Trudeau LE, Yue Z, Krainc D, Oertel W, Obeso JA, Volpicelli-Daley L. Neuronal vulnerability in Parkinson disease: Should the focus be on axons and synaptic terminals? Mov Disord 2019; 34:1406-1422. [PMID: 31483900 PMCID: PMC6879792 DOI: 10.1002/mds.27823] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/03/2019] [Accepted: 07/12/2019] [Indexed: 12/11/2022] Open
Abstract
While current effective therapies are available for the symptomatic control of PD, treatments to halt the progressive neurodegeneration still do not exist. Loss of dopamine neurons in the SNc and dopamine terminals in the striatum drive the motor features of PD. Multiple lines of research point to several pathways which may contribute to dopaminergic neurodegeneration. These pathways include extensive axonal arborization, mitochondrial dysfunction, dopamine's biochemical properties, abnormal protein accumulation of α-synuclein, defective autophagy and lysosomal degradation, and synaptic impairment. Thus, understanding the essential features and mechanisms of dopaminergic neuronal vulnerability is a major scientific challenge and highlights an outstanding need for fostering effective therapies against neurodegeneration in PD. This article, which arose from the Movement Disorders 2018 Conference, discusses and reviews the possible mechanisms underlying neuronal vulnerability and potential therapeutic approaches in PD. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yvette Wong
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Philadelphia, PA, 19104-4283, USA
| | - Kerry Purtell
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Hess Research Center 9th Floor, New York, NY 10029, USA
| | - Samuel Burke Nanni
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - A. Jon Stoessl
- University of British Columbia and Vancouver Coastal Health, Pacific Parkinson’s Research Centre & National Parkinson Foundation Centre of Excellence, 2221 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Louis-Eric Trudeau
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Hess Research Center 9th Floor, New York, NY 10029, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Wolfgang Oertel
- Department of Neurology, Philipps University Marburg, Baldingerstraße 1, 35043, Marburg, Germany
| | - Jose A. Obeso
- HM CINAC, HM Puerta del Sur, Hospitales de Madrid, Mostoles Medical School, CEU-San Pablo University, and CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Laura Volpicelli-Daley
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
43
|
Dong C, Garen CR, Mercier P, Petersen NO, Woodside MT. Characterizing the inhibition of α-synuclein oligomerization by a pharmacological chaperone that prevents prion formation by the protein PrP. Protein Sci 2019; 28:1690-1702. [PMID: 31306510 DOI: 10.1002/pro.3684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
Abstract
Aggregation of the disordered protein α-synuclein into amyloid fibrils is a central feature of synucleinopathies, neurodegenerative disorders that include Parkinson's disease. Small, pre-fibrillar oligomers of misfolded α-synuclein are thought to be the key toxic entities, and α-synuclein misfolding can propagate in a prion-like way. We explored whether a compound with anti-prion activity that can bind to unfolded parts of the protein PrP, the cyclic tetrapyrrole Fe-TMPyP, was also active against α-synuclein aggregation. Observing the initial stages of aggregation via fluorescence cross-correlation spectroscopy, we found that Fe-TMPyP inhibited small oligomer formation in a dose-dependent manner. Fe-TMPyP also inhibited the formation of mature amyloid fibrils in vitro, as detected by thioflavin T fluorescence. Isothermal titration calorimetry indicated Fe-TMPyP bound to monomeric α-synuclein with a stoichiometry of 2, and two-dimensional heteronuclear single quantum coherence NMR spectra revealed significant interactions between Fe-TMPyP and the C-terminus of the protein. These results suggest commonalities among aggregation mechanisms for α-synuclein and the prion protein may exist that can be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Chunhua Dong
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Craig R Garen
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Pascal Mercier
- National High Field Nuclear Magnetic Resonance Centre (NANUC), Edmonton, Alberta, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
44
|
Savitt D, Jankovic J. Targeting α-Synuclein in Parkinson's Disease: Progress Towards the Development of Disease-Modifying Therapeutics. Drugs 2019; 79:797-810. [PMID: 30982161 DOI: 10.1007/s40265-019-01104-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative movement disorder, is characterized by progressive motor and non-motor symptoms [1]. Despite treatment with pharmacologic and surgical therapies, the disease will continue to relentlessly advance. Hence, there is a great deal of interest in potential disease-modifying therapies with the hope that the neurodegenerative process can be slowed or halted. The purpose of this review is to highlight the role toxic α-synuclein (α-syn) plays in PD pathogenesis and critically review the relevant literature about therapeutic modalities targeting α-syn. Toxic α-syn plays a key role in PD pathogenesis, disrupting important cellular functions, and, thus, targeting α-syn is a reasonable disease-modifying strategy. Current approaches under investigation include decreasing α-syn production with RNA interference (RNAi), inhibiting α-syn aggregation, promoting intracellular degradation of α-syn aggregates (via enhancing autophagy and enhancing lysosomal degradation), and promoting extracellular degradation of α-syn via active and passive immunization.
Collapse
Affiliation(s)
- Daniel Savitt
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX, 77030-4202, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX, 77030-4202, USA.
| |
Collapse
|
45
|
Killinger BA, Kordower JH. Spreading of alpha-synuclein - relevant or epiphenomenon? J Neurochem 2019; 150:605-611. [PMID: 31152606 DOI: 10.1111/jnc.14779] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 01/10/2023]
Abstract
The intracellular accumulation of misfolded alpha-synuclein pathology, termed Lewy pathology, throughout the brain is a phenomenon central to Parkinson's disease pathogenesis. In recent years it has become apparent that Lewy pathology can spread from neuron-to-neuron and between interconnected brain regions. Understanding the phenomenon of Lewy pathology propagation holds great promise in its explanatory power to determine the etiology of Parkinson's disease and related synucleinopathies. However, it remains to be seen if the spread of Lewy pathology is critical for driving this disease. Here we discuss the spreading of Lewy pathology while highlighting some important concepts and experimental observations. We conclude that further studies are required to determine if, and how, the spreading behavior of Lewy pathology is involved in Parkinson's disease. "This article is part of the Special Issue Synuclein".
Collapse
Affiliation(s)
- Bryan A Killinger
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
46
|
Froula JM, Castellana-Cruz M, Anabtawi NM, Camino JD, Chen SW, Thrasher DR, Freire J, Yazdi AA, Fleming S, Dobson CM, Kumita JR, Cremades N, Volpicelli-Daley LA. Defining α-synuclein species responsible for Parkinson's disease phenotypes in mice. J Biol Chem 2019; 294:10392-10406. [PMID: 31142553 DOI: 10.1074/jbc.ra119.007743] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by fibrillar neuronal inclusions composed of aggregated α-synuclein (α-syn). These inclusions are associated with behavioral and pathological PD phenotypes. One strategy for therapeutic interventions is to prevent the formation of these inclusions to halt disease progression. α-Synuclein exists in multiple structural forms, including disordered, nonamyloid oligomers, ordered amyloid oligomers, and fibrils. It is critical to understand which conformers contribute to specific PD phenotypes. Here, we utilized a mouse model to explore the pathological effects of stable β-amyloid-sheet oligomers compared with those of fibrillar α-synuclein. We biophysically characterized these species with transmission EM, atomic-force microscopy, CD spectroscopy, FTIR spectroscopy, analytical ultracentrifugation, and thioflavin T assays. We then injected these different α-synuclein forms into the mouse striatum to determine their ability to induce PD-related phenotypes. We found that β-sheet oligomers produce a small but significant loss of dopamine neurons in the substantia nigra pars compacta (SNc). Injection of small β-sheet fibril fragments, however, produced the most robust phenotypes, including reduction of striatal dopamine terminals, SNc loss of dopamine neurons, and motor-behavior defects. We conclude that although the β-sheet oligomers cause some toxicity, the potent effects of the short fibrillar fragments can be attributed to their ability to recruit monomeric α-synuclein and spread in vivo and hence contribute to the development of PD-like phenotypes. These results suggest that strategies to reduce the formation and propagation of β-sheet fibrillar species could be an important route for therapeutic intervention in PD and related disorders.
Collapse
Affiliation(s)
- Jessica M Froula
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Marta Castellana-Cruz
- the Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom, and
| | - Nadia M Anabtawi
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - José D Camino
- the Institute for Biocomputation and Physics of Complex Systems (BIFI)-Joint Unit BIFI-IQFR (CSIC), University of Zaragoza, Zaragoza 50018, Spain
| | - Serene W Chen
- the Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom, and
| | - Drake R Thrasher
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jennifer Freire
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Allen A Yazdi
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Sheila Fleming
- the Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Christopher M Dobson
- the Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom, and
| | - Janet R Kumita
- the Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom, and
| | - Nunilo Cremades
- the Institute for Biocomputation and Physics of Complex Systems (BIFI)-Joint Unit BIFI-IQFR (CSIC), University of Zaragoza, Zaragoza 50018, Spain,
| | - Laura A Volpicelli-Daley
- From the Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294,
| |
Collapse
|
47
|
Flores-Cuadrado A, Saiz-Sanchez D, Mohedano-Moriano A, Martinez-Marcos A, Ubeda-Bañon I. Neurodegeneration and contralateral α-synuclein induction after intracerebral α-synuclein injections in the anterior olfactory nucleus of a Parkinson's disease A53T mouse model. Acta Neuropathol Commun 2019; 7:56. [PMID: 30987677 PMCID: PMC6463651 DOI: 10.1186/s40478-019-0713-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease is characterized by a proteinopathy that includes aggregates of α-synuclein. A recent hypothesis proposes a prion-like spreading mechanism for this α-synucleinopathy. Early neuropathological deposits occur, among others, in the anterior olfactory nucleus (AON). This study investigates the anterograde and/or retrograde transmissibility of exogenous α-synuclein inoculated in the right AON of the A53T model of Parkinson’s disease and wild-type mice as well as neuronal and glial involvement. Seven experimental groups were established: wild-type injected with tracers; A53T mice injected with either α-synuclein or saline 2 months beforehand; wild-type injected with either α-synuclein or saline 2 months beforehand; and wild-type injected with either α-synuclein or saline 4 months beforehand. Weight and behavioral changes were analyzed. Immunohistochemistry against α-synuclein, NeuN, Iba-1 and GFAP was performed. Volume and marker distributions in the olfactory bulb (OB), AON and piriform cortex were analyzed using unbiased stereology. The behavioral analyses reveal higher levels of hyperactivity in transgenic as compared to wild-type mice. Tract-tracing experiments show that the main contralateral afferent projections to the dorsal AON come from the AON and secondarily from the OB. In saline-injected transgenic animals, α-synuclein expression in the OB and the AON is higher in the left hemisphere than in the right hemisphere, which could be due to basal interhemispheric differences. α-synuclein injection could provoke a significant increase in the left hemisphere of the transgenic mice’s OB, compared to saline-injected animals. Neuronal loss was observed in saline-injected transgenic mice relative to the saline-injected wild-type group. There were no overall differences in neuron number following injection of α-synuclein into either wild-type or transgenic mice, however some neuron loss was apparent in specific regions of α-synuclein injected wild-types. Microglia labeling appeared to be correlated with surgery-induced inflammation. Astroglial labeling was higher in transgenic animals, which could be due to endogenous α-synucleinopathy. This study suggests α-synucleinopathy induction, via retrograde and contralateral projections, within the olfactory system of transgenic animals.
Collapse
|
48
|
Tremblay ME, Cookson MR, Civiero L. Glial phagocytic clearance in Parkinson's disease. Mol Neurodegener 2019; 14:16. [PMID: 30953527 PMCID: PMC6451240 DOI: 10.1186/s13024-019-0314-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/15/2019] [Indexed: 12/21/2022] Open
Abstract
An emerging picture suggests that glial cells' loss of beneficial roles or gain of toxic functions can contribute to neurodegenerative conditions. Among glial cells, microglia and astrocytes have been shown to play phagocytic roles by engulfing synapses, apoptotic cells, cell debris, and released toxic proteins. As pathogenic protein accumulation is a key feature in Parkinson's disease (PD), compromised phagocytic clearance might participate in PD pathogenesis. In contrast, enhanced, uncontrolled and potentially toxic glial clearance capacity could contribute to synaptic degeneration. Here, we summarize the current knowledge of the molecular mechanisms underlying microglial and astrocytic phagocytosis, focusing on the possible implication of phagocytic dysfunction in neuronal degeneration. Several endo-lysosomal proteins displaying genetic variants in PD are highly expressed by microglia and astrocytes. We also present the evidence that lysosomal defects can affect phagocytic clearance and discuss the therapeutic relevance of restoring or enhancing lysosomal function in PD.
Collapse
Affiliation(s)
- Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC Canada
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC Canada
| | - Mark R. Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD USA
| | - Laura Civiero
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| |
Collapse
|
49
|
Geisler JG. 2,4 Dinitrophenol as Medicine. Cells 2019; 8:cells8030280. [PMID: 30909602 PMCID: PMC6468406 DOI: 10.3390/cells8030280] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
In the sanctity of pure drug discovery, objective reasoning can become clouded when pursuing ideas that appear unorthodox, but are spot on physiologically. To put this into historical perspective, it was an unorthodox idea in the 1950’s to suggest that warfarin, a rat poison, could be repositioned into a breakthrough drug in humans to protect against strokes as a blood thinner. Yet it was approved in 1954 as Coumadin® and has been prescribed to billions of patients as a standard of care. Similarly, no one can forget the horrific effects of thalidomide, prescribed or available without a prescription, as both a sleeping pill and “morning sickness” anti-nausea medication targeting pregnant women in the 1950’s. The “thalidomide babies” became the case-in-point for the need of strict guidelines by the U.S. Food & Drug Administration (FDA) or full multi-species teratogenicity testing before drug approval. More recently it was found that thalidomide is useful in graft versus host disease, leprosy and resistant tuberculosis treatment, and as an anti-angiogenesis agent as a breakthrough drug for multiple myeloma (except for pregnant female patients). Decades of diabetes drug discovery research has historically focused on every possible angle, except, the energy-out side of the equation, namely, raising mitochondrial energy expenditure with chemical uncouplers. The idea of “social responsibility” allowed energy-in agents to be explored and the portfolio is robust with medicines of insulin sensitizers, insulin analogues, secretagogues, SGLT2 inhibitors, etc., but not energy-out medicines. The primary reason? It appeared unorthodox, to return to exploring a drug platform used in the 1930s in over 100,000 obese patients used for weight loss. This is over 80-years ago and prior to Dr Peter Mitchell explaining the mechanism of how mitochondrial uncouplers, like 2,4-dinitrophenol (DNP) even worked by three decades later in 1961. Although there is a clear application for metabolic disease, it was not until recently that this platform was explored for its merit at very low, weight-neutral doses, for treating insidious human illnesses and completely unrelated to weight reduction. It is known that mitochondrial uncouplers specifically target the entire organelle’s physiology non-genomically. It has been known for years that many neuromuscular and neurodegenerative diseases are associated with overt production of reactive oxygen species (ROSs), a rise in isoprostanes (biomarker of mitochondrial ROSs in urine or blood) and poor calcium (Ca2+) handing. It has also been known that mitochondrial uncouplers lower ROS production and Ca2+ overload. There is evidence that elevation of isoprostanes precedes disease onset, in Alzheimer’s Disease (AD). It is also curious, why so many neurodegenerative diseases of known and unknown etiology start at mid-life or later, such as Multiple Sclerosis (MS), Huntington Disease (HD), AD, Parkinson Disease, and Amyotrophic Lateral Sclerosis (ALS). Is there a relationship to a buildup of mutations that are sequestered over time due to ROSs exceeding the rate of repair? If ROS production were managed, could disease onset due to aging be delayed or prevented? Is it possible that most, if not all neurodegenerative diseases are manifested through mitochondrial dysfunction? Although DNP, a historic mitochondrial uncoupler, was used in the 1930s at high doses for obesity in well over 100,000 humans, and so far, it has never been an FDA-approved drug. This review will focus on the application of using DNP, but now, repositioned as a potential disease-modifying drug for a legion of insidious diseases at much lower and paradoxically, weight neutral doses. DNP will be addressed as a treatment for “metabesity”, an emerging term related to the global comorbidities associated with the over-nutritional phenotype; obesity, diabetes, nonalcoholic steatohepatitis (NASH), metabolic syndrome, cardiovascular disease, but including neurodegenerative disorders and accelerated aging. Some unexpected drug findings will be discussed, such as DNP’s induction of neurotrophic growth factors involved in neuronal heath, learning and cognition. For the first time in 80’s years, the FDA has granted (to Mitochon Pharmaceutical, Inc., Blue Bell, PA, USA) an open Investigational New Drug (IND) approval to begin rigorous clinical testing of DNP for safety and tolerability, including for the first ever, pharmacokinetic profiling in humans. Successful completion of Phase I clinical trial will open the door to explore the merits of DNP as a possible treatment of people with many truly unmet medical needs, including those suffering from HD, MS, PD, AD, ALS, Duchenne Muscular Dystrophy (DMD), and Traumatic Brain Injury (TBI).
Collapse
Affiliation(s)
- John G Geisler
- Mitochon Pharmaceuticals, Inc., 970 Cross Lane, Blue Bell, PA 19422, USA.
| |
Collapse
|
50
|
Brundin P, Coetzee GA. Genetically engineered stem cell-derived neurons can be rendered resistant to alpha-synuclein aggregate pathology. Eur J Neurosci 2019; 49:316-319. [PMID: 30614081 DOI: 10.1111/ejn.14333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| | - Gerhard A Coetzee
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|