1
|
Fleming SM, Scott S, Hamad EJ, Herman DE, Holden JG, Yan L, Linning-Duffy K, Kemp CJ, Patterson JR, Miller KM, Kubik M, Kuhn N, Stoll AC, Duffy MF, Steece-Collier K, Cole-Strauss A, Lipton JW, Luk KC, Sortwell CE. Intrastriatal injection of alpha-synuclein preformed fibrils to rats results in L-DOPA reversible sensorimotor impairments and alterations in non-motor function. Front Neurosci 2025; 19:1556447. [PMID: 40236948 PMCID: PMC11996896 DOI: 10.3389/fnins.2025.1556447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction The alpha-synuclein (α-syn) preformed fibril (PFF) model of Parkinson's disease (PD) is widely used in rodents to understand the mechanisms contributing to progression of pathology and neurodegeneration in the disorder. While the time course of pathology in the α-syn PFF rat model has been well characterized, it has been more challenging to determine reliable and reproducible behavior impairments. This is mainly due to α-syn PFF injections resulting in a partial nigrostriatal lesion that make motor anomalies more subtle and difficult to detect, just as in patients with PD. In the present study we sought to examine the effect of increased striatal distribution and injection quantity of α-syn PFFs in rats on accumulation of phosphorylated α-syn inclusions, nigrostriatal degeneration, sensorimotor behavior, and nonmotor function related to PD. Methods Male Fischer 344 rats were injected unilaterally in the striatum with a total of 24μg α-syn PFFs distributed into three sites, or an equal volume of phosphate buffered saline (PBS) as a control condition. Sensorimotor function was assessed using a battery of behavioral tests sensitive to varying degrees of nigrostriatal neurodegeneration. Non-motor testing included assays for olfaction, emotional reactivity, cognitive function, and sleep. Results At six months post injection, α-syn PFF rats displayed significant movement and somatosensory asymmetries compared with control rats. Time to initiate a forelimb step and time to contact an adhesive stimulus on the forepaw took significantly longer with the contralateral limb compared with the ipsilateral limb in α-syn PFF rats. Further, hindlimb stepping in the cylinder was significantly reduced in α-syn PFF-injected rats compared with controls. Cognitive function was also affected in the α-syn PFF rats, with investigation time significantly decreased in an object recognition test. Levodopa reversibility was observed in the movement initiation and cylinder tests. Postmortem analysis revealed a 55% loss of nigral tyrosine hydroxylase immunoreactive neurons and a 63% reduction in striatal dopamine content in α-syn PFF-injected rats. Conclusion Thus, using the present α-syn PFF surgical parameters, sufficient nigrostriatal degeneration can be achieved to manifest significant motor and non-motor deficits. These rat α-syn PFF surgical parameters will be important for preclinical assessment of novel diseasemodifying therapies.
Collapse
Affiliation(s)
- Sheila M. Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sophia Scott
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Edward J. Hamad
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Danielle E. Herman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - John G. Holden
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Christopher J. Kemp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Joseph R. Patterson
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathryn M. Miller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Nathan Kuhn
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Anna C. Stoll
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Megan F. Duffy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Jack W. Lipton
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kelvin C. Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Caryl E. Sortwell
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| |
Collapse
|
2
|
Sampson TR, Tansey MG, West AB, Liddle RA. Lewy body diseases and the gut. Mol Neurodegener 2025; 20:14. [PMID: 39885558 PMCID: PMC11783828 DOI: 10.1186/s13024-025-00804-5] [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: 10/17/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Gastrointestinal (GI) involvement in Lewy body diseases (LBDs) has been observed since the initial descriptions of patients by James Parkinson. Recent experimental and human observational studies raise the possibility that pathogenic alpha-synuclein (⍺-syn) might develop in the GI tract and subsequently spread to susceptible brain regions. The cellular and mechanistic origins of ⍺-syn propagation in disease are under intense investigation. Experimental LBD models have implicated important contributions from the intrinsic gut microbiome, the intestinal immune system, and environmental toxicants, acting as triggers and modifiers to GI pathologies. Here, we review the primary clinical observations that link GI dysfunctions to LBDs. We first provide an overview of GI anatomy and the cellular repertoire relevant for disease, with a focus on luminal-sensing cells of the intestinal epithelium including enteroendocrine cells that express ⍺-syn and make direct contact with nerves. We describe interactions within the GI tract with resident microbes and exogenous toxicants, and how these may directly contribute to ⍺-syn pathology along with related metabolic and immunological responses. Finally, critical knowledge gaps in the field are highlighted, focusing on pivotal questions that remain some 200 years after the first descriptions of GI tract dysfunction in LBDs. We predict that a better understanding of how pathophysiologies in the gut influence disease risk and progression will accelerate discoveries that will lead to a deeper overall mechanistic understanding of disease and potential therapeutic strategies targeting the gut-brain axis to delay, arrest, or prevent disease progression.
Collapse
Affiliation(s)
- Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30329, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Malú Gámez Tansey
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Normal Fixel Institute of Neurological Diseases, Gainesville, FL, 32608, USA
| | - Andrew B West
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Center for Neurodegeneration and Neurotherapeutic Research, Department of Pharmacology and Cancer Biology, Durham, NC, 27710, USA.
| | - Rodger A Liddle
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA.
- Department of Medicine, Duke University and Department of Veterans Affairs Health Care System, Durham, NC, 27710, USA.
| |
Collapse
|
3
|
Trist BG, Wright CJ, Rangel A, Cottle L, Prasad A, Jensen NM, Gram H, Dzamko N, Jensen PH, Kirik D. Novel tools to quantify total, phospho-Ser129 and aggregated alpha-synuclein in the mouse brain. NPJ Parkinsons Dis 2024; 10:217. [PMID: 39516469 PMCID: PMC11549080 DOI: 10.1038/s41531-024-00830-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Assays for quantifying aggregated and phosphorylated (S129) human α-synuclein protein are widely used to evaluate pathological burden in patients suffering from synucleinopathy disorders. Many of these assays, however, do not cross-react with mouse α-synuclein or exhibit poor sensitivity for this target, which is problematic considering the preponderance of mouse models at the forefront of pre-clinical α-synuclein research. In this project, we addressed this unmet need by reformulating two existing AlphaLISA® SureFire® Ultra™ total and pS129 α-synuclein assay kits to yield robust and ultrasensitive (LLoQ ≤ 0.5 pg/mL) quantification of mouse and human wild-type and pS129 α-synuclein protein. We then employed these assays, together with the BioLegend α-synuclein aggregate ELISA, to assess α-synuclein S129 phosphorylation and aggregation in different mouse brain tissue preparations. Overall, we highlight the compatibility of these new immunoassays with rodent models and demonstrate their potential to advance knowledge surrounding α-synuclein phosphorylation and aggregation in synucleinopathies.
Collapse
Affiliation(s)
- Benjamin Guy Trist
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
| | - Courtney Jade Wright
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Brain Repair and Imaging in Neural Systems (BRAINS), Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alejandra Rangel
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
| | - Louise Cottle
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Asheeta Prasad
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Nanna Møller Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Hjalte Gram
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Nicolas Dzamko
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Poul Henning Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Deniz Kirik
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Brain Repair and Imaging in Neural Systems (BRAINS), Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
4
|
Sokratian A, Zhou Y, Tatli M, Burbidge KJ, Xu E, Viverette E, Donzelli S, Duda AM, Yuan Y, Li H, Strader S, Patel N, Shiell L, Malankhanova T, Chen O, Mazzulli JR, Perera L, Stahlberg H, Borgnia M, Bartesaghi A, Lashuel HA, West AB. Mouse α-synuclein fibrils are structurally and functionally distinct from human fibrils associated with Lewy body diseases. SCIENCE ADVANCES 2024; 10:eadq3539. [PMID: 39485845 PMCID: PMC11800946 DOI: 10.1126/sciadv.adq3539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024]
Abstract
The intricate process of α-synuclein aggregation and fibrillization holds pivotal roles in Parkinson's disease (PD) and multiple system atrophy (MSA). While mouse α-synuclein can fibrillize in vitro, whether these fibrils commonly used in research to induce this process or form can reproduce structures in the human brain remains unknown. Here, we report the first atomic structure of mouse α-synuclein fibrils, which was solved in parallel by two independent teams. The structure shows striking similarity to MSA-amplified and PD-associated E46K fibrils. However, mouse α-synuclein fibrils display altered packing arrangements, reduced hydrophobicity, and heightened fragmentation sensitivity and evoke only weak immunological responses. Furthermore, mouse α-synuclein fibrils exhibit exacerbated pathological spread in neurons and humanized α-synuclein mice. These findings provide critical insights into the structural underpinnings of α-synuclein pathogenicity and emphasize a need to reassess the role of mouse α-synuclein fibrils in the development of related diagnostic probes and therapeutic interventions.
Collapse
Affiliation(s)
- Arpine Sokratian
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Ye Zhou
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Meltem Tatli
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, and Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Kevin J. Burbidge
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Enquan Xu
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Elizabeth Viverette
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Sonia Donzelli
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Addison M. Duda
- Department of Chemistry, Duke University, Durham, NC 27708, USA
| | - Yuan Yuan
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Huizhong Li
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Samuel Strader
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Nirali Patel
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Lauren Shiell
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Tuyana Malankhanova
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Olivia Chen
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Joseph R. Mazzulli
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lalith Perera
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, and Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Mario Borgnia
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Alberto Bartesaghi
- Department of Computer Science, Duke University, Durham, NC 27708, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27705, USA
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27708, USA
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Qatar Foundation ND BioSciences, Qatar Foundation Headquarters, PO Box 3400, Al Rayyan, Qatar
| | - Andrew B. West
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
5
|
Massari CM, Dues DJ, Bergsma A, Sipple K, Frye M, Williams ET, Moore DJ. Neuropathology in an α-synuclein preformed fibril mouse model occurs independent of the Parkinson's disease-linked lysosomal ATP13A2 protein. Neurobiol Dis 2024; 202:106701. [PMID: 39406291 DOI: 10.1016/j.nbd.2024.106701] [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: 08/08/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Loss-of-function mutations in the ATP13A2 (PARK9) gene are implicated in early-onset autosomal recessive Parkinson's disease (PD) and other neurodegenerative disorders. ATP13A2 encodes a lysosomal transmembrane P5B-type ATPase that is highly expressed in brain and specifically within the substantia nigra pars compacta (SNc). Recent studies have revealed its normal role as a lysosomal polyamine transporter, although its contribution to PD-related pathology remains unclear. Cellular studies report that ATP13A2 can regulate α-synuclein (α-syn) secretion via exosomes. However, the relationship between ATP13A2 and α-syn in animal models remains inconclusive. ATP13A2 knockout (KO) mice exhibit lysosomal abnormalities and reactive astrogliosis but do not develop PD-related neuropathology. Studies manipulating α-syn levels in mice lacking ATP13A2 indicate minimal effects on pathology. The delivery of α-syn preformed fibrils (PFFs) into the mouse striatum is a well-defined model to study the development and spread of α-syn pathology. In this study we unilaterally injected wild-type (WT) and homozygous ATP13A2 KO mice with mouse α-syn PFFs in the striatum and evaluated mice for neuropathology after 6 months. The distribution, spread and extent of α-syn aggregation in multiple regions of the mouse brain was largely independent of ATP13A2 expression. The loss of nigrostriatal pathway dopaminergic neurons and their nerve terminals induced by PFFs were equivalent in WT and ATP13A2 KO mice. Reactive astrogliosis was induced equivalently by α-syn PFFs in WT and KO mice but was already significantly higher in ATP13A2 KO mice due to pre-existing reactive gliosis. We did not identify asymmetric motor disturbances, microglial activation, or axonal damage induced by α-syn PFFs in WT or KO mice. Although α-syn PFFs induce an increase in lysosomal number in the SNc in general, TH-positive dopaminergic neurons did not exhibit either increased lysosomal area or intensity, regardless of genotype. Our study evaluating the spread of α-syn pathology reveals no exacerbation of α-syn pathology, neuronal loss, astrogliosis or motor deficits in ATP13A2 KO mice, suggesting that selective lysosomal abnormalities resulting from ATP13A2 loss do not play a major role in α-syn clearance or propagation in vivo.
Collapse
Affiliation(s)
- Caio M Massari
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Dylan J Dues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Alexis Bergsma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Kayla Sipple
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Maxwell Frye
- West Michigan Neurodegenerative Diseases (MiND) Program, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Erin T Williams
- West Michigan Neurodegenerative Diseases (MiND) Program, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
6
|
Guimarães RP, de Resende MCS, Tavares MM, Belardinelli de Azevedo C, Ruiz MCM, Mortari MR. Construct, Face, and Predictive Validity of Parkinson's Disease Rodent Models. Int J Mol Sci 2024; 25:8971. [PMID: 39201659 PMCID: PMC11354451 DOI: 10.3390/ijms25168971] [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: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally. Current drugs only alleviate symptoms without halting disease progression, making rodent models essential for researching new therapies and understanding the disease better. However, selecting the right model is challenging due to the numerous models and protocols available. Key factors in model selection include construct, face, and predictive validity. Construct validity ensures the model replicates pathological changes seen in human PD, focusing on dopaminergic neurodegeneration and a-synuclein aggregation. Face validity ensures the model's symptoms mirror those in humans, primarily reproducing motor and non-motor symptoms. Predictive validity assesses if treatment responses in animals will reflect those in humans, typically involving classical pharmacotherapies and surgical procedures. This review highlights the primary characteristics of PD and how these characteristics are validated experimentally according to the three criteria. Additionally, it serves as a valuable tool for researchers in selecting the most appropriate animal model based on established validation criteria.
Collapse
Affiliation(s)
- Rayanne Poletti Guimarães
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Maria Clara Souza de Resende
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Mesquita Tavares
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Caio Belardinelli de Azevedo
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Cesar Merino Ruiz
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
- Neurological Rehabilitation Unit, Sarah Network of Rehabilitation Hospitals, Brasília 70335-901, Brazil
| | - Márcia Renata Mortari
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| |
Collapse
|
7
|
Laursen ALS, Olesen MV, Folke J, Brudek T, Knecht LH, Sotty F, Lambertsen KL, Fog K, Dalgaard LT, Aznar S. Systemic inflammation activates coagulation and immune cell infiltration pathways in brains with propagating α-synuclein fibril aggregates. Mol Cell Neurosci 2024; 129:103931. [PMID: 38508542 DOI: 10.1016/j.mcn.2024.103931] [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: 01/04/2024] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
Synucleinopathies are a group of diseases characterized by brain aggregates of α-synuclein (α-syn). The gradual accumulation of α-syn and the role of inflammation in early-stage pathogenesis remain poorly understood. We explored this interaction by inducing chronic inflammation in a common pre-clinical synucleinopathy mouse model. Three weeks post unilateral intra-striatal injections of human α-syn pre-formed fibrils (PFF), mice underwent repeated intraperitoneal injections of 1 mg/ml lipopolysaccharide (LPS) for 3 weeks. Histological examinations of the ipsilateral site showed phospho-α-syn regional spread and LPS-induced neutrophil recruitment to the brain vasculature. Biochemical assessment of the contralateral site confirmed spreading of α-syn aggregation to frontal cortex and a rise in intracerebral TNF-α, IL-1β, IL-10 and KC/GRO cytokines levels due to LPS. No LPS-induced exacerbation of α-syn pathology load was observed at this stage. Proteomic analysis was performed contralateral to the PFF injection site using LC-MS/MS. Subsequent downstream Reactome Gene-Set Analysis indicated that α-syn pathology alters mitochondrial metabolism and synaptic signaling. Chronic LPS-induced inflammation further lead to an overrepresentation of pathways related to fibrin clotting as well as integrin and B cell receptor signaling. Western blotting confirmed a PFF-induced increase in fibrinogen brain levels and a PFF + LPS increase in Iba1 levels, indicating activated microglia. Splenocyte profiling revealed changes in T and B cells, monocytes, and neutrophils populations due to LPS treatment in PFF injected animals. In summary, early α-syn pathology impacts energy homeostasis pathways, synaptic signaling and brain fibrinogen levels. Concurrent mild systemic inflammation may prime brain immune pathways in interaction with peripheral immunity.
Collapse
Affiliation(s)
- Anne-Line Strange Laursen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark; Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Mikkel Vestergaard Olesen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Jonas Folke
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Tomasz Brudek
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Luisa Harriet Knecht
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløwsvej 21-25, DK-5000, Odense, Denmark; Department of Neurology, Odense University Hospital, J.B. Winsløwsvej 4, Odense, Denmark; BRIDGE - Brain-Research-Inter-Disciplinary Guided Excellence, Department of Clinical Institute, University of Southern Denmark, Winsløwparken 19, Odense, Denmark.
| | - Karina Fog
- H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark.
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Susana Aznar
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| |
Collapse
|
8
|
Soto I, McManus R, Navarrete W, Kasanga EA, Doshier K, Nejtek VA, Salvatore MF. Aging accelerates locomotor decline in PINK1 knockout rats in association with decreased nigral, but not striatal, dopamine and tyrosine hydroxylase expression. Exp Neurol 2024; 376:114771. [PMID: 38580154 DOI: 10.1016/j.expneurol.2024.114771] [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: 02/01/2024] [Revised: 03/15/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Parkinson's disease (PD) rodent models provide insight into the relationship between nigrostriatal dopamine (DA) signaling and locomotor function. Although toxin-based rat models produce frank nigrostriatal neuron loss and eventual motor decline characteristic of PD, the rapid nature of neuronal loss may not adequately translate premotor traits, such as cognitive decline. Unfortunately, rodent genetic PD models, like the Pink1 knockout (KO) rat, often fail to replicate the differential severity of striatal DA and tyrosine hydroxylase (TH) loss, and a bradykinetic phenotype, reminiscent of human PD. To elucidate this inconsistency, we evaluated aging as a progression factor in the timing of motor and non-motor cognitive impairments. Male PINK1 KO and age-matched wild type (WT) rats were evaluated in a longitudinal study from 3 to 16 months old in one cohort, and in a cross-sectional study of young adult (6-7 months) and aged (18-19 months) in another cohort. Young adult PINK1 KO rats exhibited hyperkinetic behavior associated with elevated DA and TH in the substantia nigra (SN), which decreased therein, but not striatum, in the aged KO rats. Additionally, norepinephrine levels decreased in aged KO rats in the prefrontal cortex (PFC), paired with a higher DA levels in young and aged KO. Although a younger age of onset characterizes familial forms of PD, our results underscore the critical need to consider age-related factors. Moreover, the results indicate that compensatory mechanisms may exist to preserve locomotor function, evidenced by increased DA in the SN early in the lifespan, in response to deficient PINK1 function, which declines with aging and the onset of motor decline.
Collapse
Affiliation(s)
- Isabel Soto
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Robert McManus
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Walter Navarrete
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Ella A Kasanga
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Kirby Doshier
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Vicki A Nejtek
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Michael F Salvatore
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America.
| |
Collapse
|
9
|
Vaidya B, Gupta P, Biswas S, Laha JK, Roy I, Sharma SS. Effect of Clemizole on Alpha-Synuclein-Preformed Fibrils-Induced Parkinson's Disease Pathology: A Pharmacological Investigation. Neuromolecular Med 2024; 26:19. [PMID: 38703217 DOI: 10.1007/s12017-024-08785-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with mitochondrial dysfunctions and oxidative stress. However, to date, therapeutics targeting these pathological events have not managed to translate from bench to bedside for clinical use. One of the major reasons for the lack of translational success has been the use of classical model systems that do not replicate the disease pathology and progression with the same degree of robustness. Therefore, we employed a more physiologically relevant model involving alpha-synuclein-preformed fibrils (PFF) exposure to SH-SY5Y cells and Sprague Dawley rats. We further explored the possible involvement of transient receptor potential canonical 5 (TRPC5) channels in PD-like pathology induced by these alpha-synuclein-preformed fibrils with emphasis on amelioration of oxidative stress and mitochondrial health. We observed that alpha-synuclein PFF exposure produced neurobehavioural deficits that were positively ameliorated after treatment with the TRPC5 inhibitor clemizole. Furthermore, Clemizole also reduced p-alpha-synuclein and diminished oxidative stress levels which resulted in overall improvements in mitochondrial biogenesis and functions. Finally, the results of the pharmacological modulation were further validated using siRNA-mediated knockdown of TRPC5 channels, which also decreased p-alpha-synuclein expression. Together, the results of this study could be superimposed in the future for exploring the beneficial effects of TRPC5 channel modulation for other neurodegenerative disorders and synucleopathies.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pankaj Gupta
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Mohali, Punjab, 160062, India
| | - Soumojit Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Joydev K Laha
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Mohali, Punjab, 160062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
10
|
Wang T, Liu W, Zhang Q, Jiao J, Wang Z, Gao G, Yang H. 4-Oxo-2-Nonenal- and Agitation-Induced Aggregates of α-Synuclein and Phosphorylated α-Synuclein with Distinct Biophysical Properties and Biomedical Applications. Cells 2024; 13:739. [PMID: 38727274 PMCID: PMC11082957 DOI: 10.3390/cells13090739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
α-Synuclein (α-syn) can form oligomers, protofibrils, and fibrils, which are associated with the pathogenesis of Parkinson's disease and other synucleinopathies. Both the lipid peroxidation product 4-oxo-2-nonenal (ONE) and agitation can induce aggregation of α-syn and phosphorylated α-syn. Thus, clarification of the characteristics of different α-syn species could help to select suitable aggregates for diagnosis and elucidate the pathogenesis of diseases. Here, we characterized ONE-induced wild-type (WT) α-syn aggregates (OW), ONE-induced phosphorylated α-syn (p-α-syn) aggregates (OP), agitation-induced α-syn preformed fibrils (PFF), and agitation-induced p-α-syn preformed fibrils (pPFF). Thioflavin T (ThT) dying demonstrated that OW and OP had fewer fibrils than the PFF and pPFF. Transmission electron microscopy revealed that the lengths of PFF and pPFF were similar, but the diameters differed. OW and OP had more compact structures than PFF and pPFF. Aggregation of p-α-syn was significantly faster than WT α-syn. Furthermore, OW and OP were more sodium dodecyl sulfate-stable and proteinase K-resistant, suggesting greater stability and compactness, while aggregates of PFF and pPFF were more sensitive to proteinase K treatment. Both ONE- and agitation-induced aggregates were cytotoxic when added exogenously to SH-SY5Y cells with increasing incubation times, but the agitation-induced aggregates caused cell toxicity in a shorter time and more p-α-syn inclusions. Similarly, p-proteins were more cytotoxic than non-p-proteins. Finally, all four aggregates were used as standard antigens to establish sandwich enzyme-linked immunosorbent assay (ELISA). The results showed that the recognition efficiency of OW and OP was more sensitive than that of PFF and pPFF. The OW- and OP-specific ELISA for detection of p-α-syn and α-syn in plasma samples of Thy1-α-syn transgenic mice showed that the content of aggregates could reflect the extent of disease. ONE and agitation induced the formation of α-syn aggregates with distinct biophysical properties and biomedical applications.
Collapse
Affiliation(s)
- Tie Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Weijin Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Qidi Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Jie Jiao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Zihao Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Ge Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| | - Hui Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (T.W.); (W.L.); (Q.Z.); (J.J.); (Z.W.)
- Center of Parkinson’s Disease, Beijing Key Laboratory of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Institute of Brain Disorders, Beijing 100069, China
| |
Collapse
|
11
|
Melachroinou K, Divolis G, Tsafaras G, Karampetsou M, Fortis S, Stratoulias Y, Papadopoulou G, Kriebardis AG, Samiotaki M, Vekrellis K. Endogenous Alpha-Synuclein is Essential for the Transfer of Pathology by Exosome-Enriched Extracellular Vesicles, Following Inoculation with Preformed Fibrils in vivo. Aging Dis 2024; 15:869-892. [PMID: 37548944 PMCID: PMC10917543 DOI: 10.14336/ad.2023.0614] [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: 04/07/2023] [Accepted: 06/14/2023] [Indexed: 08/08/2023] Open
Abstract
The main pathological hallmark of Parkinson's disease (PD) and related synucleinopathies is the presence of intracellular proteinaceous aggregates, enriched in the presynaptic protein alpha-Synuclein (α-Syn). α-Syn association with exosomes has been previously documented both as a physiological process of secretion and as a pathological process of disease transmission, however, critical information about the mechanisms governing this interplay is still lacking. To address this, we utilized the α-Syn preformed fibril (PFF) mouse model of PD, as a source of brain-derived exosome-enriched extracellular vesicles (ExE-EVs) and assessed their pathogenic capacity following intrastriatal injections in host wild type (WT) mouse brain. We further investigated the impact of the fibrillar α-Syn on the exosomal cargo independent of the endogenous α-Syn, by isolating ExE-EVs from PFF-injected α-Syn knockout mice. Although PFF inoculation does not alter the morphology, size distribution, and quantity of brain-derived ExE-EVs, it triggers changes in the exosomal proteome related to synaptic and mitochondrial function, as well as metabolic processes. Importantly, we showed that the presence of the endogenous α-Syn is essential for the ExE-EVs to acquire a pathogenic capacity, allowing them to mediate disease transmission by inducing phosphorylated-α-Syn pathology. Notably, misfolded α-Syn containing ExE-EVs when injected in WT mice were able to induce astrogliosis and synaptic alterations in the host brain, at very early stages of α-Syn pathology, preceding the formation of the insoluble α-Syn accumulations. Collectively, our data suggest that exosomal cargo defines their ability to spread α-Syn pathology.
Collapse
Affiliation(s)
- Katerina Melachroinou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Georgios Divolis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - George Tsafaras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Mantia Karampetsou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Sotirios Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece.
| | - Yannis Stratoulias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Gina Papadopoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Kostas Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
12
|
Lv QK, Tao KX, Yao XY, Pang MZ, Cao BE, Liu CF, Wang F. Melatonin MT1 receptors regulate the Sirt1/Nrf2/Ho-1/Gpx4 pathway to prevent α-synuclein-induced ferroptosis in Parkinson's disease. J Pineal Res 2024; 76:e12948. [PMID: 38488331 DOI: 10.1111/jpi.12948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/29/2024] [Accepted: 03/02/2024] [Indexed: 03/19/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic (DA) neurons and aggregation of α-synuclein (α-syn). Ferroptosis, a form of cell death induced by iron accumulation and lipid peroxidation, is involved in the pathogenesis of PD. It is unknown whether melatonin receptor 1 (MT1) modulates α-syn and ferroptosis in PD. Here, we used α-syn preformed fibrils (PFFs) to induce PD models in vivo and in vitro. In PD mice, α-syn aggregation led to increased iron deposition and ferroptosis. MT1 knockout exacerbated these changes and resulted in more DA neuronal loss and severe motor impairment. MT1 knockout also suppressed the Sirt1/Nrf2/Ho1/Gpx4 pathway, reducing resistance to ferroptosis, and inhibited expression of ferritin Fth1, leading to more release of ferrous ions. In vitro experiments confirmed these findings. Knockdown of MT1 enhanced α-syn PFF-induced intracellular α-syn aggregation and suppressed expression of the Sirt1/Nrf2/Ho1/Gpx4 pathway and Fth1 protein, thereby aggravating ferroptosis. Conversely, overexpression of MT1 reversed these effects. Our findings reveal a novel mechanism by which MT1 activation prevents α-syn-induced ferroptosis in PD, highlighting the neuroprotective role of MT1 in PD.
Collapse
Affiliation(s)
- Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Kang-Xin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiao-Yu Yao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Zhu Pang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bing-Er Cao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Soto I, McManus R, Navarrete-Barahona W, Kasanga EA, Doshier K, Nejtek VA, Salvatore MF. Aging hastens locomotor decline in PINK1 knockout rats in association with decreased nigral, but not striatal, dopamine and tyrosine hydroxylase expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578317. [PMID: 38352365 PMCID: PMC10862808 DOI: 10.1101/2024.02.01.578317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Parkinson's disease (PD) rodent models provide insight into the relationship between nigrostriatal dopamine (DA) signaling and locomotor function. Although toxin-based rat models produce frank nigrostriatal neuron loss and eventual motor decline characteristic of PD, the rapid nature of neuronal loss may not adequately translate premotor traits, such as cognitive decline. Unfortunately, rodent genetic PD models, like the Pink1 knockout (KO) rat, often fail to replicate the differential severity of striatal DA and tyrosine hydroxylase (TH) loss, and a bradykinetic phenotype, reminiscent of human PD. To elucidate this inconsistency, we evaluated aging as a progression factor in the timing of motor and non-motor cognitive impairments. Male PINK1 KO and age-matched wild type (WT) rats were evaluated in a longitudinal study from 3 to 16 months old in one cohort, and in a cross-sectional study of young adult (6-7 months) and aged (18-19 months) in another cohort. Young adult PINK1 KO rats exhibited hyperkinetic behavior associated with elevated DA and TH in the substantia nigra (SN), which decreased therein, but not striatum, in the aged KO rats. Additionally, norepinephrine levels decreased in aged KO rats in the prefrontal cortex (PFC), paired with a higher DA content in young and aged KO. Although a younger age of onset characterizes familial forms of PD, our results underscore the critical need to consider age-related factors. Moreover, the results indicate that compensatory mechanisms may exist to preserve locomotor function, evidenced by increased DA in the SN early in the lifespan, in response to deficient PINK1 function, which declines with aging and the onset of motor impairment.
Collapse
|
14
|
Dorion MF, Yaqubi M, Senkevich K, Kieran NW, MacDonald A, Chen CXQ, Luo W, Wallis A, Shlaifer I, Hall JA, Dudley RWR, Glass IA, Stratton JA, Fon EA, Bartels T, Antel JP, Gan-or Z, Durcan TM, Healy LM. MerTK is a mediator of alpha-synuclein fibril uptake by human microglia. Brain 2024; 147:427-443. [PMID: 37671615 PMCID: PMC10834256 DOI: 10.1093/brain/awad298] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/26/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Mer tyrosine kinase (MerTK) is a receptor tyrosine kinase that mediates non-inflammatory, homeostatic phagocytosis of diverse types of cellular debris. Highly expressed on the surface of microglial cells, MerTK is of importance in brain development, homeostasis, plasticity and disease. Yet, involvement of this receptor in the clearance of protein aggregates that accumulate with ageing and in neurodegenerative diseases has yet to be defined. The current study explored the function of MerTK in the microglial uptake of alpha-synuclein fibrils which play a causative role in the pathobiology of synucleinopathies. Using human primary and induced pluripotent stem cell-derived microglia, the MerTK-dependence of alpha-synuclein fibril internalization was investigated in vitro. Relevance of this pathway in synucleinopathies was assessed through burden analysis of MERTK variants and analysis of MerTK expression in patient-derived cells and tissues. Pharmacological inhibition of MerTK and siRNA-mediated MERTK knockdown both caused a decreased rate of alpha-synuclein fibril internalization by human microglia. Consistent with the non-inflammatory nature of MerTK-mediated phagocytosis, alpha-synuclein fibril internalization was not observed to induce secretion of pro-inflammatory cytokines such as IL-6 or TNF, and downmodulated IL-1β secretion from microglia. Burden analysis in two independent patient cohorts revealed a significant association between rare functionally deleterious MERTK variants and Parkinson's disease in one of the cohorts (P = 0.002). Despite a small upregulation in MERTK mRNA expression in nigral microglia from Parkinson's disease/Lewy body dementia patients compared to those from non-neurological control donors in a single-nuclei RNA-sequencing dataset (P = 5.08 × 10-21), no significant upregulation in MerTK protein expression was observed in human cortex and substantia nigra lysates from Lewy body dementia patients compared to controls. Taken together, our findings define a novel role for MerTK in mediating the uptake of alpha-synuclein fibrils by human microglia, with possible involvement in limiting alpha-synuclein spread in synucleinopathies such as Parkinson's disease. Upregulation of this pathway in synucleinopathies could have therapeutic values in enhancing alpha-synuclein fibril clearance in the brain.
Collapse
Affiliation(s)
- Marie-France Dorion
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Konstantin Senkevich
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montreal H3A 0C7, Canada
| | - Nicholas W Kieran
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Adam MacDonald
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Carol X Q Chen
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Wen Luo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Amber Wallis
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Jeffery A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Roy W R Dudley
- Department of Pediatric Surgery, Division of Neurosurgery, Montreal Children's Hospital, McGill University Health Centers, Montreal H4A 3J1, Canada
| | - Ian A Glass
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Edward A Fon
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Tim Bartels
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Ziv Gan-or
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montreal H3A 0C7, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| |
Collapse
|
15
|
Boyton I, Valenzuela SM, Collins-Praino LE, Care A. Neuronanomedicine for Alzheimer's and Parkinson's disease: Current progress and a guide to improve clinical translation. Brain Behav Immun 2024; 115:631-651. [PMID: 37967664 DOI: 10.1016/j.bbi.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 09/19/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023] Open
Abstract
Neuronanomedicine is an emerging multidisciplinary field that aims to create innovative nanotechnologies to treat major neurodegenerative disorders, such as Alzheimer's (AD) and Parkinson's disease (PD). A key component of neuronanomedicine are nanoparticles, which can improve drug properties and demonstrate enhanced safety and delivery across the blood-brain barrier, a major improvement on existing therapeutic approaches. In this review, we critically analyze the latest nanoparticle-based strategies to modify underlying disease pathology to slow or halt AD/PD progression. We find that a major roadblock for neuronanomedicine translation to date is a poor understanding of how nanoparticles interact with biological systems (i.e., bio-nano interactions), which is partly due to inconsistent reporting in published works. Accordingly, this review makes a set of specific recommendations to help guide researchers to harness the unique properties of nanoparticles and thus realise breakthrough treatments for AD/PD.
Collapse
Affiliation(s)
- India Boyton
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | | | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia.
| |
Collapse
|
16
|
Ferreira SA, Li C, Klæstrup IH, Vitic Z, Rasmussen RK, Kirkegaard A, Toft GU, Betzer C, Svendsen P, Jensen PH, Luo Y, Etzerodt A, Moestrup SK, Romero-Ramos M. Sex-dimorphic neuroprotective effect of CD163 in an α-synuclein mouse model of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:164. [PMID: 38092806 PMCID: PMC10719342 DOI: 10.1038/s41531-023-00606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Alpha-synuclein (α-syn) aggregation and immune activation represent hallmark pathological events in Parkinson's disease (PD). The PD-associated immune response encompasses both brain and peripheral immune cells, although little is known about the immune proteins relevant for such a response. We propose that the upregulation of CD163 observed in blood monocytes and in the responsive microglia in PD patients is a protective mechanism in the disease. To investigate this, we used the PD model based on intrastriatal injections of murine α-syn pre-formed fibrils in CD163 knockout (KO) mice and wild-type littermates. CD163KO females revealed an impaired and differential early immune response to α-syn pathology as revealed by immunohistochemical and transcriptomic analysis. After 6 months, CD163KO females showed an exacerbated immune response and α-syn pathology, which ultimately led to dopaminergic neurodegeneration of greater magnitude. These findings support a sex-dimorphic neuroprotective role for CD163 during α-syn-induced neurodegeneration.
Collapse
Affiliation(s)
- Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Conghui Li
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ida H Klæstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Zagorka Vitic
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | | | - Asger Kirkegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Gitte U Toft
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Pia Svendsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren K Moestrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
17
|
Alwani A, Maziarz K, Burda G, Jankowska-Kiełtyka M, Roman A, Łyszczarz G, Er S, Barut J, Barczyk-Woźnicka O, Pyza E, Kreiner G, Nalepa I, Chmielarz P. Investigating the potential effects of α-synuclein aggregation on susceptibility to chronic stress in a mouse Parkinson's disease model. Pharmacol Rep 2023; 75:1474-1487. [PMID: 37725330 PMCID: PMC10661792 DOI: 10.1007/s43440-023-00530-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a motor disorder characterized by the degeneration of dopaminergic neurons, putatively due to the accumulation of α-synuclein (α-syn) in Lewy bodies (LBs) in Substantia Nigra. PD is also associated with the formation of LBs in brain areas responsible for emotional and cognitive regulation such as the amygdala and prefrontal cortex, and concurrent depression prevalence in PD patients. The exact link between dopaminergic cell loss, α-syn aggregation, depression, and stress, a major depression risk factor, is unclear. Therefore, we aimed to explore the interplay between sensitivity to chronic stress and α-syn aggregation. METHODS Bilateral injections of α-syn preformed fibrils (PFFs) into the striatum of C57Bl/6 J mice were used to induce α-syn aggregation. Three months after injections, animals were exposed to chronic social defeat stress. RESULTS α-syn aggregation did not affect stress susceptibility but independently caused increased locomotor activity in the open field test, reduced anxiety in the light-dark box test, and increased active time in the tail suspension test. Ex vivo analysis revealed modest dopaminergic neuron loss in the substantia nigra and reduced dopaminergic innervation in the dorsal striatum in PFFs injected groups. α-Syn aggregates were prominent in the amygdala, prefrontal cortex, and substantia nigra, with minimal α-syn aggregation in the raphe nuclei and locus coeruleus. CONCLUSIONS Progressive bilateral α-syn aggregation might lead to compensatory activity increase and alterations in emotionally regulated behavior, without affecting stress susceptibility. Understanding how α-syn aggregation and degeneration in specific brain structures contribute to depression and anxiety in PD patients requires further investigation.
Collapse
Affiliation(s)
- Anna Alwani
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Katarzyna Maziarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Gabriela Burda
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Monika Jankowska-Kiełtyka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Adam Roman
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Gabriela Łyszczarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Safak Er
- Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Justyna Barut
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Olga Barczyk-Woźnicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
18
|
Kasanga EA, Han Y, Shifflet MK, Navarrete W, McManus R, Parry C, Barahona A, Nejtek VA, Manfredsson FP, Kordower JH, Richardson JR, Salvatore MF. Nigral-specific increase in ser31 phosphorylation compensates for tyrosine hydroxylase protein and nigrostriatal neuron loss: Implications for delaying parkinsonian signs. Exp Neurol 2023; 368:114509. [PMID: 37634696 DOI: 10.1016/j.expneurol.2023.114509] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Compensatory mechanisms that augment dopamine (DA) signaling are thought to mitigate onset of hypokinesia prior to major loss of tyrosine hydroxylase (TH) in striatum that occurs in Parkinson's disease. However, the identity of such mechanisms remains elusive. In the present study, the rat nigrostriatal pathway was unilaterally-lesioned with 6-hydroxydopamine (6-OHDA) to determine whether differences in DA content, TH protein, TH phosphorylation, or D1 receptor expression in striatum or substantia nigra (SN) aligned with hypokinesia onset and severity at two time points. In striatum, DA and TH loss reached its maximum (>90%) 7 days after lesion induction. However, in SN, no DA loss occurred, despite ∼60% TH loss. Hypokinesia was established at 21 days post-lesion and maintained at 28 days. At this time, DA loss was ∼60% in the SN, but still of lesser magnitude than TH loss. At day 7 and 28, ser31 TH phosphorylation increased only in SN, corresponding to less DA versus TH protein loss. In contrast, ser40 TH phosphorylation was unaffected in either region. Despite DA loss in both regions at day 28, D1 receptor expression increased only in lesioned SN. These results support the concept that augmented components of DA signaling in the SN, through increased ser31 TH phosphorylation and D1 receptor expression, contribute as compensatory mechanisms against progressive nigrostriatal neuron and TH protein loss, and may mitigate hypokinesia severity.
Collapse
Affiliation(s)
- Ella A Kasanga
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Yoonhee Han
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Marla K Shifflet
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Walter Navarrete
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Robert McManus
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Caleb Parry
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Arturo Barahona
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Vicki A Nejtek
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85287, USA
| | - Jason R Richardson
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Michael F Salvatore
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA.
| |
Collapse
|
19
|
Zhang S, Dauer K, Strohäker T, Tatenhorst L, Caldi Gomes L, Mayer S, Jung BC, Kim WS, Lee S, Becker S, Liesche‐Starnecker F, Zweckstetter M, Lingor P. Alpha-synuclein fibrils amplified from multiple system atrophy and Parkinson's disease patient brain spread after intracerebral injection into mouse brain. Brain Pathol 2023; 33:e13196. [PMID: 37485772 PMCID: PMC10467043 DOI: 10.1111/bpa.13196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB) are neurodegenerative disorders with alpha-synuclein (α-syn) aggregation pathology. Different strains of α-syn with unique properties are suggested to cause distinct clinical and pathological manifestations resulting in PD, MSA, or DLB. To study individual α-syn spreading patterns, we injected α-syn fibrils amplified from brain homogenates of two MSA patients and two PD patients into the brains of C57BI6/J mice. Antibody staining against pS129-α-syn showed that α-syn fibrils amplified from the brain homogenates of the four different patients caused different levels of α-syn spreading. The strongest α-syn pathology was triggered by α-syn fibrils of one of the two MSA patients, followed by comparable pS129-α-syn induction by the second MSA and one PD patient material. Histological analysis using an antibody against Iba1 further showed that the formation of pS129-α-syn is associated with increased microglia activation. In contrast, no differences in dopaminergic neuron numbers or co-localization of α-syn in oligodendrocytes were observed between the different groups. Our data support the spreading of α-syn pathology in MSA, while at the same time pointing to spreading heterogeneity between different patients potentially driven by individual patient immanent factors.
Collapse
Affiliation(s)
- Shuyu Zhang
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Karina Dauer
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Timo Strohäker
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Lars Tatenhorst
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Lucas Caldi Gomes
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Simon Mayer
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Byung Chul Jung
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Woojin S. Kim
- Faculty of Medicine and Health, Brain and Mind Centre and School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesUniversity of New South Wales and Neuroscience Research AustraliaRandwickNew South WalesAustralia
| | - Seung‐Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Stefan Becker
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Friederike Liesche‐Starnecker
- Department of Neuropathology, Institute of Pathology, School of MedicineTechnical University MunichMunichGermany
- Department of Pathology and Molecular Diagnostics, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Paul Lingor
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
20
|
Dues DJ, Nguyen APT, Becker K, Ma J, Moore DJ. Hippocampal subfield vulnerability to α-synuclein pathology precedes neurodegeneration and cognitive dysfunction. NPJ Parkinsons Dis 2023; 9:125. [PMID: 37640722 PMCID: PMC10462636 DOI: 10.1038/s41531-023-00574-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Cognitive dysfunction is a salient feature of Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). The onset of dementia reflects the spread of Lewy pathology throughout forebrain structures. The mere presence of Lewy pathology, however, provides limited indication of cognitive status. Thus, it remains unclear whether Lewy pathology is the de facto substrate driving cognitive dysfunction in PD and DLB. Through application of α-synuclein fibrils in vivo, we sought to examine the influence of pathologic inclusions on cognition. Following stereotactic injection of α-synuclein fibrils within the mouse forebrain, we measured the burden of α-synuclein pathology at 1-, 3-, and 6-months post-injection within subregions of the hippocampus and cortex. Under this paradigm, the hippocampal CA2/3 subfield was especially susceptible to α-synuclein pathology. Strikingly, we observed a drastic reduction of pathology in the CA2/3 subfield across time-points, consistent with the consolidation of α-synuclein pathology into dense somatic inclusions followed by neurodegeneration. Silver-positive degenerating neurites were observed prior to neuronal loss, suggesting that this might be an early feature of fibril-induced neurotoxicity and a precursor to neurodegeneration. Critically, mice injected with α-synuclein fibrils developed progressive deficits in spatial learning and memory. These findings support that the formation of α-synuclein inclusions in the mouse forebrain precipitate neurodegenerative changes that recapitulate features of Lewy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Dylan J Dues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - An Phu Tran Nguyen
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Katelyn Becker
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jiyan Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Chinese Institute for Brain Research, Beijing, China
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
21
|
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
|
22
|
Guo Q, Kawahata I, Jia W, Wang H, Cheng A, Yabuki Y, Shioda N, Fukunaga K. α-Synuclein decoy peptide protects mice against α-synuclein-induced memory loss. CNS Neurosci Ther 2023; 29:1547-1560. [PMID: 36786129 PMCID: PMC10173724 DOI: 10.1111/cns.14120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
AIMS We previously found that a decoy peptide derived from the C-terminal sequence of α-Synuclein (αSyn) prevents cytotoxic αSyn aggregation caused by fatty acid-binding protein 3 (FABP3) in vitro. In this study, we continued to utilize αSyn-derived peptides to further validate their effects on αSyn neurotoxicity and behavioral impairments in αSyn preformed fibrils (PFFs)-injected mouse model of Parkinson's disease (PD). METHODS Mice were injected with αSyn PFFs in the bilateral olfactory bulb (OB) and then were subjected to behavioral analysis at 2-week intervals post-injection. Peptides nasal administration was initiated one week after injection. Changes in phosphorylation of αSyn and neuronal damage in the OB were measured using immunostaining at week 4. The effect of peptides on the interaction between αSyn and FABP3 was examined using co-immunoprecipitation. RESULTS αSyn PFF-injected mice showed significant memory loss but no motor function impairment. Long-term nasal treatment with peptides effectively prevented memory impairment. In peptide-treated αSyn PFF-injected mice, the peptides entered the OB smoothly through the nasal cavity and were mainly concentrated in neurons in the mitral cell layer, significantly suppressing the excessive phosphorylation of αSyn and reducing the formation of αSyn-FABP3 oligomers, thereby preventing neuronal death. The addition of peptides also blocked the interaction of αSyn and FABP3 at the recombinant protein level, and its effect was strongest at molar concentrations comparable to those of αSyn and FABP3. CONCLUSIONS Our findings suggest that the αSyn decoy peptide represents a novel therapeutic approach for reducing the accumulation of toxic αSyn-FABP3 oligomers in the brain, thereby preventing the progression of synucleinopathies.
Collapse
Affiliation(s)
- Qingyun Guo
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wenbin Jia
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haoyang Wang
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - An Cheng
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.,BRI Pharma Incorporated, Sendai, Japan
| |
Collapse
|
23
|
Guo M, Liu W, Luo H, Shao Q, Li Y, Gu Y, Guan Y, Ma W, Chen M, Yang H, Ji X, Liu J. Hypoxic stress accelerates the propagation of pathological alpha-synuclein and degeneration of dopaminergic neurons. CNS Neurosci Ther 2022; 29:544-558. [PMID: 36514210 PMCID: PMC9873519 DOI: 10.1111/cns.14055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/12/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS The etiology of Parkinson's disease (PD) is complex and the mechanism is unclear. It has become a top priority to find common factors that induce and affect PD pathology. We explored the key role of hypoxia in promoting the pathological propagation of α-synuclein (α-syn) and the progression of PD. METHODS We performed PD modeling by conducting intracranial stereotaxic surgery in the unilateral striatum of mice. We then measured protein aggregation in vitro. The rotarod and pole tests were employed next to measure the damage of the phenotype. Pathological deposition and autophagy were also observed by immunofluorescence staining and protein levels measured by western blotting. RESULTS We demonstrated that short-term hypoxia activated phosphorylated (p)-α-syn in mice. We confirmed that p-α-syn was more readily formed aggregates than α-syn in vitro. Furthermore, we found that hypoxia promoted the activation and propagation of endogenous α-syn, contributing to the earlier degeneration of dopaminergic neurons in the substantia nigra and the deposition of p-α-syn in our animal model. Finally, autophagy inhibition contributed to the above pathologies. CONCLUSION Hypoxia was shown to accelerate the pathological progression and damage phenotype in PD model mice. The results provided a promising research target for determining common interventions for PD in the future.
Collapse
Affiliation(s)
- Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Weijin Liu
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina,School of Rehabilitation MedicineCapital Medical UniversityBeijingChina
| | - Hanjiang Luo
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuning Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Min Chen
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Hui Yang
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
24
|
Unique seeding profiles and prion-like propagation of synucleinopathies are highly dependent on the host in human α-synuclein transgenic mice. Acta Neuropathol 2022; 143:663-685. [PMID: 35488930 DOI: 10.1007/s00401-022-02425-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/22/2022]
Abstract
α-synuclein (αSyn) is an intrinsically disordered protein which can undergo structural transformations, resulting in the formation of stable, insoluble fibrils. αSyn amyloid-type nucleation can be induced by misfolded 'seeds' serving as a conformational template, tantamount to the prion-like mechanism. Accumulation of αSyn inclusions is a key feature of dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), and are found as additional pathology in Alzheimer's disease (AD) such as AD with amygdala predominant Lewy bodies (AD/ALB). While these disorders accumulate the same pathological protein, they exhibit heterogeneity in clinical and histological features; however, the mechanism(s) underlying this variability remains elusive. Accruing data from human autopsy studies, animal inoculation modeling, and in vitro characterization experiments, have lent credence to the hypothesis that conformational polymorphism of the αSyn amyloid-type fibril structure results in distinct "strains" with categorical infectivity traits. Herein, we directly compare the seeding abilities and outcome of human brain lysates from these diseases, as well as recombinant preformed human αSyn fibrils by the intracerebral inoculation of transgenic mice overexpressing either human wild-type αSyn or human αSyn with the familial A53T mutation. Our study has revealed that the initiating inoculum heavily dictates the phenotypic and pathological course of disease. Interestingly, we have also established relevant host-dependent distinctions between propagation profiles, including burden and spread of inclusion pathology throughout the neuroaxis, as well as severity of neurological symptoms. These findings provide compelling evidence supporting the hypothesis that diverse prion-type conformers may explain the variability seen in synucleinopathies.
Collapse
|