51
|
Zhang Y, Zhang Y, Mao C, Jiang Z, Fan G, Wang E, Chen Y, Palaniyappan L. Association of Cortical Gyrification With Imaging and Serum Biomarkers in Patients With Parkinson Disease. Neurology 2023; 101:e311-e323. [PMID: 37268433 PMCID: PMC10382266 DOI: 10.1212/wnl.0000000000207410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/30/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Pathologic progression across the cortex is a key feature of Parkinson disease (PD). Cortical gyrification is a morphologic feature of human cerebral cortex that is tightly linked to the integrity of underlying axonal connectivity. Monitoring cortical gyrification reductions may provide a sensitive marker of progression through structural connectivity, preceding the progressive stages of PD pathology. We aimed to examine the progressive cortical gyrification reductions and their associations with overlying cortical thickness, white matter (WM) integrity, striatum dopamine availability, serum neurofilament light (NfL) chain, and CSF α-synuclein levels in PD. METHODS This study included a longitudinal dataset with baseline (T0), 1-year (T1), and 4-year (T4) follow-ups and 2 cross-sectional datasets. Local gyrification index (LGI) was computed from T1-weighted MRI data to measure cortical gyrification. Fractional anisotropy (FA) was computed from diffusion-weighted MRI data to measure WM integrity. Striatal binding ratio (SBR) was measured from 123Ioflupane SPECT scans. Serum NfL and CSF α-synuclein levels were also measured. RESULTS The longitudinal dataset included 113 patients with de novo PD and 55 healthy controls (HCs). The cross-sectional datasets included 116 patients with relatively more advanced PD and 85 HCs. Compared with HCs, patients with de novo PD showed accelerated LGI and FA reductions over 1-year period and a further decline at 4-year follow-up. Across the 3 time points, the LGI paralleled and correlated with FA (p = 0.002 at T0, p = 0.0214 at T1, and p = 0.0037 at T4) and SBR (p = 0.0095 at T0, p = 0.0035 at T1, and p = 0.0096 at T4) but not with overlying cortical thickness in patients with PD. Both LGI and FA correlated with serum NfL level (LGI: p < 0.0001 at T0, p = 0.0043 at T1; FA: p < 0.0001 at T0, p = 0.0001 at T1) but not with CSF α-synuclein level in patients with PD. In the 2 cross-sectional datasets, we revealed similar patterns of LGI and FA reductions and associations between LGI and FA in patients with more advanced PD. DISCUSSION We demonstrated progressive reductions in cortical gyrification that were robustly associated with WM microstructure, striatum dopamine availability, and serum NfL level in PD. Our findings may contribute biomarkers for PD progression and potential pathways for early interventions of PD.
Collapse
Affiliation(s)
- Yuanchao Zhang
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada.
| | - Yu Zhang
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada.
| | - Chengjie Mao
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada
| | - Zhen Jiang
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada
| | - Guohua Fan
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada
| | - Erlei Wang
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada.
| | - Yifan Chen
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada
| | - Lena Palaniyappan
- From the School of Life Science and Technology (Yuanchao Zhang, Y.C.), University of Electronic Science and Technology of China, Chengdu, Sichuan; Artificial Intelligence Research Institute (Yu Zhang), Zhejiang Lab, Hangzhou; Department of Neurology (C.M.), and Department of Radiology (Z.J., G.F., E.W.), The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Douglas Mental Health University Institute (L.P.), McGill University, Montreal, Quebec, Canada
| |
Collapse
|
52
|
Kim MS, Ra EA, Kweon SH, Seo BA, Ko HS, Oh Y, Lee G. Advanced human iPSC-based preclinical model for Parkinson's disease with optogenetic alpha-synuclein aggregation. Cell Stem Cell 2023; 30:973-986.e11. [PMID: 37339636 PMCID: PMC10829432 DOI: 10.1016/j.stem.2023.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/02/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) offer advantages for disease modeling and drug discovery. However, recreating innate cellular pathologies, particularly in late-onset neurodegenerative diseases with accumulated protein aggregates including Parkinson's disease (PD), has been challenging. To overcome this barrier, we developed an optogenetics-assisted α-synuclein (α-syn) aggregation induction system (OASIS) that rapidly induces α-syn aggregates and toxicity in PD hiPSC-midbrain dopaminergic neurons and midbrain organoids. Our OASIS-based primary compound screening with SH-SY5Y cells identified 5 candidates that were secondarily validated with OASIS PD hiPSC-midbrain dopaminergic neurons and midbrain organoids, leading us to finally select BAG956. Furthermore, BAG956 significantly reverses characteristic PD phenotypes in α-syn preformed fibril models in vitro and in vivo by promoting autophagic clearance of pathological α-syn aggregates. Following the FDA Modernization Act 2.0's emphasis on alternative non-animal testing methods, our OASIS can serve as an animal-free preclinical test model (newly termed "nonclinical test") for the synucleinopathy drug development.
Collapse
Affiliation(s)
- Min Seong Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eun A Ra
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sin Ho Kweon
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bo Am Seo
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju-si, Gangwon-do, Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju-si, Gangwon-do, Korea; Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju-si, Gangwon-do, Korea
| | - Han Seok Ko
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yohan Oh
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea.
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
53
|
Mirzac D, Kreis SL, Luhmann HJ, Gonzalez-Escamilla G, Groppa S. Translating Pathological Brain Activity Primers in Parkinson's Disease Research. RESEARCH (WASHINGTON, D.C.) 2023; 6:0183. [PMID: 37383218 PMCID: PMC10298229 DOI: 10.34133/research.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Translational experimental approaches that help us better trace Parkinson's disease (PD) pathophysiological mechanisms leading to new therapeutic targets are urgently needed. In this article, we review recent experimental and clinical studies addressing abnormal neuronal activity and pathological network oscillations, as well as their underlying mechanisms and modulation. Our aim is to enhance our knowledge about the progression of Parkinson's disease pathology and the timing of its symptom's manifestation. Here, we present mechanistic insights relevant for the generation of aberrant oscillatory activity within the cortico-basal ganglia circuits. We summarize recent achievements extrapolated from available PD animal models, discuss their advantages and limitations, debate on their differential applicability, and suggest approaches for transferring knowledge on disease pathology into future research and clinical applications.
Collapse
Affiliation(s)
- Daniela Mirzac
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Svenja L. Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sergiu Groppa
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
54
|
Chatterjee D, Krainc D. Mechanisms of Glucocerebrosidase Dysfunction in Parkinson's Disease. J Mol Biol 2023; 435:168023. [PMID: 36828270 PMCID: PMC10247409 DOI: 10.1016/j.jmb.2023.168023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Beta-glucocerebrosidase is a lysosomal hydrolase, encoded by GBA1 that represents the most common risk gene associated with Parkinson's disease (PD) and Lewy Body Dementia. Glucocerebrosidase dysfunction has been also observed in the absence of GBA1 mutations across different genetic and sporadic forms of PD and related disorders, suggesting a broader role of glucocerebrosidase in neurodegeneration. In this review, we highlight recent advances in mechanistic characterization of glucocerebrosidase function as the foundation for development of novel therapeutics targeting glucocerebrosidase in PD and related disorders.
Collapse
Affiliation(s)
- Diptaman Chatterjee
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA. https://twitter.com/NeilChatterBox
| | - Dimitri Krainc
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
55
|
Ramirez DM, Whitesell JD, Bhagwat N, Thomas TL, Ajay AD, Nawaby A, Delatour B, Bay S, LaFaye P, Knox JE, Harris JA, Meeks JP, Diamond MI. Endogenous pathology in tauopathy mice progresses via brain networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541792. [PMID: 37293074 PMCID: PMC10245958 DOI: 10.1101/2023.05.23.541792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Neurodegenerative tauopathies are hypothesized to propagate via brain networks. This is uncertain because we have lacked precise network resolution of pathology. We therefore developed whole-brain staining methods with anti-p-tau nanobodies and imaged in 3D PS19 tauopathy mice, which have pan-neuronal expression of full-length human tau containing the P301S mutation. We analyzed patterns of p-tau deposition across established brain networks at multiple ages, testing the relationship between structural connectivity and patterns of progressive pathology. We identified core regions with early tau deposition, and used network propagation modeling to determine the link between tau pathology and connectivity strength. We discovered a bias towards retrograde network-based propagation of tau. This novel approach establishes a fundamental role for brain networks in tau propagation, with implications for human disease.
Collapse
Affiliation(s)
- Denise M.O. Ramirez
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Jennifer D. Whitesell
- Allen Institute for Brain Science; Seattle, WA, USA
- Cajal Neuroscience; Seattle, WA, USA
| | - Nikhil Bhagwat
- Allen Institute for Brain Science; Seattle, WA, USA
- McConnell Brain Imaging Centre, The Neuro (Montreal Neurological Institute-Hospital), McGill University; Montreal, Quebec, Canada
| | - Talitha L. Thomas
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Apoorva D. Ajay
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Ariana Nawaby
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Benoît Delatour
- Paris Brain Institute (ICM), CNRS UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière; Paris, France
| | - Sylvie Bay
- Unité de Chimie des Biomolécules, Institut Pasteur, Université Paris Cité, CNRS UMR 3523; Paris, France
| | - Pierre LaFaye
- Antibody Engineering Platform, Institut Pasteur, Université Paris Cité, CNRS UMR 3528; Paris, France
| | | | | | - Julian P. Meeks
- Department of Neuroscience, University of Rochester Medical School; Rochester, NY, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| |
Collapse
|
56
|
Pérez-Acuña D, Rhee KH, Shin SJ, Ahn J, Lee JY, Lee SJ. Retina-to-brain spreading of α-synuclein after intravitreal injection of preformed fibrils. Acta Neuropathol Commun 2023; 11:83. [PMID: 37210559 PMCID: PMC10199563 DOI: 10.1186/s40478-023-01575-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/22/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the aggregation of misfolded α-synuclein and progressive spreading of the aggregates from a few discrete regions to wider brain regions. Although PD has been classically considered a movement disorder, a large body of clinical evidence has revealed the progressive occurrence of non-motor symptoms. Patients present visual symptoms in the initial stages of the disease, and accumulation of phospho-α-synuclein, dopaminergic neuronal loss, and retinal thinning has been observed in the retinas of PD patients. Based on such human data, we hypothesized that α-synuclein aggregation can initiate in the retina and spread to the brain through the visual pathway. Here, we demonstrate accumulation of α-synuclein in the retinas and brains of naive mice after intravitreal injection of α-synuclein preformed fibrils (PFFs). Histological analyses showed deposition of phospho-α-synuclein inclusions within the retina 2 months after injection, with increased oxidative stress leading to loss of retinal ganglion cells and dopaminergic dysfunction. In addition, we found accumulation of phospho-α-synuclein in cortical areas with accompanying neuroinflammation after 5 months. Collectively, our findings suggest that retinal synucleinopathy lesions initiated by intravitreal injection of α-synuclein PFFs spread to various brain regions through the visual pathway in mice.
Collapse
Affiliation(s)
- Dayana Pérez-Acuña
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
| | - Ka Hyun Rhee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Soo Jean Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
| | - Jeeyun Ahn
- Department of Ophthalmology, College of Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University, Seoul, South Korea
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, South Korea.
- Neuramedy, Seoul, South Korea.
| |
Collapse
|
57
|
Carceles-Cordon M, Weintraub D, Chen-Plotkin AS. Cognitive heterogeneity in Parkinson's disease: A mechanistic view. Neuron 2023; 111:1531-1546. [PMID: 37028431 PMCID: PMC10198897 DOI: 10.1016/j.neuron.2023.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/22/2022] [Accepted: 03/13/2023] [Indexed: 04/09/2023]
Abstract
Cognitive impairment occurs in most individuals with Parkinson's disease (PD), exacting a high toll on patients, their caregivers, and the healthcare system. In this review, we begin by summarizing the current clinical landscape surrounding cognition in PD. We then discuss how cognitive impairment and dementia may develop in PD based on the spread of the pathological protein alpha-synuclein (aSyn) from neurons in brainstem regions to those in the cortical regions of the brain responsible for higher cognitive functions, as first proposed in the Braak hypothesis. We appraise the Braak hypothesis from molecular (conformations of aSyn), cell biological (cell-to-cell spread of pathological aSyn), and organ-level (region-to-region spread of aSyn pathology at the whole brain level) viewpoints. Finally, we argue that individual host factors may be the most poorly understood aspect of this pathological process, accounting for substantial heterogeneity in the pattern and pace of cognitive decline in PD.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dan Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
58
|
Nim S, O'Hara DM, Corbi-Verge C, Perez-Riba A, Fujisawa K, Kapadia M, Chau H, Albanese F, Pawar G, De Snoo ML, Ngana SG, Kim J, El-Agnaf OMA, Rennella E, Kay LE, Kalia SK, Kalia LV, Kim PM. Disrupting the α-synuclein-ESCRT interaction with a peptide inhibitor mitigates neurodegeneration in preclinical models of Parkinson's disease. Nat Commun 2023; 14:2150. [PMID: 37076542 PMCID: PMC10115881 DOI: 10.1038/s41467-023-37464-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 03/14/2023] [Indexed: 04/21/2023] Open
Abstract
Accumulation of α-synuclein into toxic oligomers or fibrils is implicated in dopaminergic neurodegeneration in Parkinson's disease. Here we performed a high-throughput, proteome-wide peptide screen to identify protein-protein interaction inhibitors that reduce α-synuclein oligomer levels and their associated cytotoxicity. We find that the most potent peptide inhibitor disrupts the direct interaction between the C-terminal region of α-synuclein and CHarged Multivesicular body Protein 2B (CHMP2B), a component of the Endosomal Sorting Complex Required for Transport-III (ESCRT-III). We show that α-synuclein impedes endolysosomal activity via this interaction, thereby inhibiting its own degradation. Conversely, the peptide inhibitor restores endolysosomal function and thereby decreases α-synuclein levels in multiple models, including female and male human cells harboring disease-causing α-synuclein mutations. Furthermore, the peptide inhibitor protects dopaminergic neurons from α-synuclein-mediated degeneration in hermaphroditic C. elegans and preclinical Parkinson's disease models using female rats. Thus, the α-synuclein-CHMP2B interaction is a potential therapeutic target for neurodegenerative disorders.
Collapse
Affiliation(s)
- Satra Nim
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Darren M O'Hara
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Carles Corbi-Verge
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Albert Perez-Riba
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Kazuko Fujisawa
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Minesh Kapadia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Hien Chau
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Federica Albanese
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Grishma Pawar
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Mitchell L De Snoo
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Sophie G Ngana
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Jisun Kim
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Omar M A El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Enrico Rennella
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lewis E Kay
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Suneil K Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada.
| | - Lorraine V Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.
| | - Philip M Kim
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
59
|
Dues DJ, Nguyen APT, Becker K, Ma J, Moore DJ. Hippocampal subfield vulnerability to α-synuclein pathology precedes neurodegeneration and cognitive dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536572. [PMID: 37090590 PMCID: PMC10120695 DOI: 10.1101/2023.04.12.536572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
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. Highlights Mice injected with α-synuclein fibrils develop hippocampal and cortical α- synuclein pathology with a dynamic regional burden at 1-, 3-, and 6-months post-injection.Silver-positive neuronal processes are an early and enduring degenerative feature of the fibril model, while extensive neurodegeneration of the hippocampal CA2/3 subfield is detected at 6-months post-injection.Mice exhibit progressive hippocampal-dependent spatial learning and memory deficits.Forebrain injection of α-synuclein fibrils may be used to model aspects of Lewy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Dylan J. Dues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - An Phu Tran Nguyen
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Katelyn Becker
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jiyan Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Darren J. Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
60
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
Collapse
|
61
|
Zhang H, D'Agostino C, Tulisiak C, Thorwald MA, Bergkvist L, Lindquist A, Meyerdirk L, Schulz E, Becker K, Steiner JA, Cacciottolo M, Kwatra M, Rey NL, Escobar Galvis ML, Ma J, Sioutas C, Morgan TE, Finch CE, Brundin P. Air pollution nanoparticle and alpha-synuclein fibrils synergistically decrease glutamate receptor A1, depending upon nPM batch activity. Heliyon 2023; 9:e15622. [PMID: 37128335 PMCID: PMC10148131 DOI: 10.1016/j.heliyon.2023.e15622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023] Open
Abstract
Background Epidemiological studies have variably linked air pollution to increased risk of Parkinson's disease (PD). However, there is little experimental evidence for this association. Alpha-synuclein (α-syn) propagation plays central roles in PD and glutamate receptor A1 (GluA1) is involved in memory and olfaction function. Methods Each mouse was exposed to one of three different batches of nano-particulate matter (nPM) (300 μg/m3, 5 h/d, 3 d/week), collected at different dates, 2017-2019, in the same urban site. After these experiments, these nPM batches were found to vary in activity. C57BL/6 female mice (3 mo) were injected with pre-formed murine α-synuclein fibrils (PFFs) (0.4 μg), which act as seeds for α-syn aggregation. Two exposure paradigms were used: in Paradigm 1, PFFs were injected into olfactory bulb (OB) prior to 4-week nPM (Batch 5b) exposure and in Paradigm 2, PFFs were injected at 4th week during 10-week nPM exposure (Batches 7 and 9). α-syn pSer129, microglia Iba1, inflammatory cytokines, and Gria1 expression were measured by immunohistochemistry or qPCR assays. Results As expected, α-syn pSer129 was detected in ipsilateral OB, anterior olfactory nucleus, amygdala and piriform cortex. One of the three batches of nPM caused a trend for elevated α-syn pSer129 in Paradigm 1, but two other batches showed no effect in Paradigm 2. However, the combination of nPM and PFF significantly decreased Gria1 mRNA in both the ipsi- and contra-lateral OB and frontal cortex for the most active two nPM batches. Neither nPM nor PFFs alone induced responses of microglia Iba1 and expression of Gria1 in the OB and cortex. Conclusion Exposures to ambient nPM had weak effect on α-syn propagation in the brain in current experimental paradigms; however, nPM and α-syn synergistically downregulated the expression of Gria1 in both OB and cortex.
Collapse
Affiliation(s)
- Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California, USA
- Corresponding author.
| | - Carla D'Agostino
- Leonard Davis School of Gerontology, University of Southern California, USA
| | | | - Max A. Thorwald
- Leonard Davis School of Gerontology, University of Southern California, USA
| | | | | | | | - Emily Schulz
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | | | | | - Mohit Kwatra
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | | | - Jiyan Ma
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Constantinos Sioutas
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, USA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, USA
| | | |
Collapse
|
62
|
Torok J, Anand C, Verma P, Raj A. Connectome-based biophysics models of Alzheimer's disease diagnosis and prognosis. Transl Res 2023; 254:13-23. [PMID: 36031051 PMCID: PMC11019890 DOI: 10.1016/j.trsl.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022]
Abstract
With the increasing prevalence of Alzheimer's disease (AD) among aging populations and the limited therapeutic options available to slow or reverse its progression, the need has never been greater for improved diagnostic tools for identifying patients in the preclinical and prodomal phases of AD. Biophysics models of the connectome-based spread of amyloid-beta (Aβ) and microtubule-associated protein tau (τ) have enjoyed recent success as tools for predicting the time course of AD-related pathological changes. However, given the complex etiology of AD, which involves not only connectome-based spread of protein pathology but also the interactions of many molecular and cellular players over multiple spatiotemporal scales, more robust, complete biophysics models are needed to better understand AD pathophysiology and ultimately provide accurate patient-specific diagnoses and prognoses. Here we discuss several areas of active research in AD whose insights can be used to enhance the mathematical modeling of AD pathology as well as recent attempts at developing improved connectome-based biophysics models. These efforts toward a comprehensive yet parsimonious mathematical description of AD hold great promise for improving both the diagnosis of patients at risk for AD and our mechanistic understanding of how AD progresses.
Collapse
Affiliation(s)
- Justin Torok
- Department of Radiology, University of California, San Francisco, San Francisco, California.
| | - Chaitali Anand
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Parul Verma
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Ashish Raj
- Department of Radiology, University of California, San Francisco, San Francisco, California; Department of Bioengineering, University of California, Berkeley and University of California, San Francisco, Berkeley, California; Department of Radiology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
63
|
Ferrari E, Salvadè M, Zianni E, Brumana M, DiLuca M, Gardoni F. Detrimental effects of soluble α-synuclein oligomers at excitatory glutamatergic synapses. Front Aging Neurosci 2023; 15:1152065. [PMID: 37009450 PMCID: PMC10060538 DOI: 10.3389/fnagi.2023.1152065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction Oligomeric and fibrillar species of the synaptic protein α-synuclein are established key players in the pathophysiology of Parkinson's disease and other synucleinopathies. Increasing evidence in the literature points to prefibrillar oligomers as the main cytotoxic species driving dysfunction in diverse neurotransmitter systems even at early disease stages. Of note, soluble oligomers have recently been shown to alter synaptic plasticity mechanisms at the glutamatergic cortico-striatal synapse. However, the molecular and morphological detrimental events triggered by soluble α-synuclein aggregates that ultimately lead to excitatory synaptic failure remain mostly elusive. Methods In the present study, we aimed to clarify the effects of soluble α-synuclein oligomers (sOligo) in the pathophysiology of synucleinopathies at cortico-striatal and hippocampal excitatory synapses. To investigate early defects of the striatal synapse in vivo, sOligo were inoculated in the dorsolateral striatum of 2-month-old wild-type C57BL/6J mice, and molecular and morphological analyses were conducted 42 and 84 days post-injection. In parallel, primary cultures of rat hippocampal neurons were exposed to sOligo, and molecular and morphological analyses were performed after 7 days of treatment. Results In vivo sOligo injection impaired the post-synaptic retention of striatal ionotropic glutamate receptors and decreased the levels of phosphorylated ERK at 84 days post-injection. These events were not correlated with morphological alterations at dendritic spines. Conversely, chronic in vitro administration of sOligo caused a significant decrease in ERK phosphorylation but did not significantly alter post-synaptic levels of ionotropic glutamate receptors or spine density in primary hippocampal neurons. Conclusion Overall, our data indicate that sOligo are involved in pathogenic molecular changes at the striatal glutamatergic synapse, confirming the detrimental effect of these species in an in vivo synucleinopathy model. Moreover, sOligo affects the ERK signaling pathway similarly in hippocampal and striatal neurons, possibly representing an early mechanism that anticipates synaptic loss.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) “Rodolfo Paoletti”, University of Milan, Milan, Italy
| |
Collapse
|
64
|
Calabresi P, Mechelli A, Natale G, Volpicelli-Daley L, Di Lazzaro G, Ghiglieri V. Alpha-synuclein in Parkinson's disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis 2023; 14:176. [PMID: 36859484 PMCID: PMC9977911 DOI: 10.1038/s41419-023-05672-9] [Citation(s) in RCA: 131] [Impact Index Per Article: 131.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/18/2023] [Accepted: 02/09/2023] [Indexed: 03/03/2023]
Abstract
Although the discovery of the critical role of α-synuclein (α-syn) in the pathogenesis of Parkinson's disease (PD) is now twenty-five years old, it still represents a milestone in PD research. Abnormal forms of α-syn trigger selective and progressive neuronal death through mitochondrial impairment, lysosomal dysfunction, and alteration of calcium homeostasis not only in PD but also in other α-syn-related neurodegenerative disorders such as dementia with Lewy bodies, multiple system atrophy, pure autonomic failure, and REM sleep behavior disorder. Furthermore, α-syn-dependent early synaptic and plastic alterations and the underlying mechanisms preceding overt neurodegeneration have attracted great interest. In particular, the presence of early inflammation in experimental models and PD patients, occurring before deposition and spreading of α-syn, suggests a mechanistic link between inflammation and synaptic dysfunction. The knowledge of these early mechanisms is of seminal importance to support the research on reliable biomarkers to precociously identify the disease and possible disease-modifying therapies targeting α-syn. In this review, we will discuss these critical issues, providing a state of the art of the role of this protein in early PD and other synucleinopathies.
Collapse
Affiliation(s)
- Paolo Calabresi
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, 00168, Italy. .,Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy.
| | - Alessandro Mechelli
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Neurologia, Università "Magna Graecia", Catanzaro, Italy
| | - Giuseppina Natale
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Laura Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Giulia Di Lazzaro
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Veronica Ghiglieri
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy.,Università Telematica San Raffaele, Rome, 00166, Italy
| |
Collapse
|
65
|
Simons E, Fleming SM. Role of rodent models in advancing precision medicine for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:3-16. [PMID: 36803818 DOI: 10.1016/b978-0-323-85555-6.00002-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson's disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.
Collapse
Affiliation(s)
- Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
66
|
Liquid-liquid Phase Separation of α-Synuclein: A New Mechanistic Insight for α-Synuclein Aggregation Associated with Parkinson's Disease Pathogenesis. J Mol Biol 2023; 435:167713. [PMID: 35787838 DOI: 10.1016/j.jmb.2022.167713] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Aberrant aggregation of the misfolded presynaptic protein, α-Synuclein (α-Syn) into Lewy body (LB) and Lewy neuritis (LN) is a major pathological hallmark of Parkinson's disease (PD) and other synucleinopathies. Numerous studies have suggested that prefibrillar and fibrillar species of the misfolded α-Syn aggregates are responsible for cell death in PD pathogenesis. However, the precise molecular events during α-Syn aggregation, especially in the early stages, remain elusive. Emerging evidence has demonstrated that liquid-liquid phase separation (LLPS) of α-Syn occurs in the nucleation step of α-Syn aggregation, which offers an alternate non-canonical aggregation pathway in the crowded microenvironment. The liquid-like α-Syn droplets gradually undergo an irreversible liquid-to-solid phase transition into amyloid-like hydrogel entrapping oligomers and fibrils. This new mechanism of α-Syn LLPS and gel formation might represent the molecular basis of cellular toxicity associated with PD. This review aims to demonstrate the recent development of α-Syn LLPS, the underlying mechanism along with the microscopic events of aberrant phase transition. This review further discusses how several intrinsic and extrinsic factors regulate the thermodynamics and kinetics of α-Syn LLPS and co-LLPS with other proteins, which might explain the pathophysiology of α-Syn in various neurodegenerative diseases.
Collapse
|
67
|
Griffin TA, Schnier PD, Cleveland EM, Newberry RW, Becker J, Carlson GA. Fibril treatment changes protein interactions of tau and α-synuclein in human neurons. J Biol Chem 2023; 299:102888. [PMID: 36634849 PMCID: PMC9978635 DOI: 10.1016/j.jbc.2023.102888] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/07/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
In several neurodegenerative disorders, the neuronal proteins tau and α-synuclein adopt aggregation-prone conformations capable of replicating within and between cells. To better understand how these conformational changes drive neuropathology, we compared the interactomes of tau and α-synuclein in the presence or the absence of recombinant fibril seeds. Human embryonic stem cells with an inducible neurogenin-2 transgene were differentiated into glutamatergic neurons expressing (1) WT 0N4R tau, (2) mutant (P301L) 0N4R tau, (3) WT α-synuclein, or (4) mutant (A53T) α-synuclein, each genetically fused to a promiscuous biotin ligase (BioID2). Neurons expressing unfused BioID2 served as controls. After treatment with fibrils or PBS, interacting proteins were labeled with biotin in situ and quantified using mass spectrometry via tandem mass tag labeling. By comparing interactions in mutant versus WT neurons and in fibril- versus PBS-treated neurons, we observed changes in protein interactions that are likely relevant to disease progression. We identified 45 shared interactors, suggesting that tau and α-synuclein function within some of the same pathways. Potential loci of shared interactions include microtubules, Wnt signaling complexes, and RNA granules. Following fibril treatment, physiological interactions decreased, whereas other interactions, including those between tau and 14-3-3 η, increased. We confirmed that 14-3-3 proteins, which are known to colocalize with protein aggregates during neurodegeneration, can promote or inhibit tau aggregation in vitro depending on the specific combination of 14-3-3 isoform and tau sequence.
Collapse
Affiliation(s)
- Tagan A Griffin
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Paul D Schnier
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Elisa M Cleveland
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Robert W Newberry
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, California, USA
| | - Julia Becker
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - George A Carlson
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, California, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, USA.
| |
Collapse
|
68
|
Peelaerts W, Baekelandt V. ⍺-Synuclein Structural Diversity and the Cellular Environment in ⍺-Synuclein Transmission Models and Humans. Neurotherapeutics 2023; 20:67-82. [PMID: 37052776 PMCID: PMC10119367 DOI: 10.1007/s13311-023-01365-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2023] [Indexed: 04/14/2023] Open
Abstract
Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) are termed synucleinopathies, disorders that are characterized by the intracellular aggregation of the protein ɑ-synuclein. The cellular tropism of synuclein pathology in these syndromes is notably distinct since in the Lewy disorders, PD and DLB, ɑSyn forms aggregates in neurons whereas in MSA ɑSyn forms aggregates in oligodendrocytes. Studies examining ɑSyn pathology in experimental models and in human brain have now identified fibrillar ɑSyn with unique but distinct molecular signatures, suggesting that the structure of these ɑSyn fibrils might be closely tied to their cellular ontogeny. In contrast to the native structural heterogeneity of ɑSyn in vitro, the conformational landscape of fibrillar ɑSyn in human brain and in vivo transmission models appears to be remarkably uniform. Here, we review the studies by which we propose a hypothesis that the cellular host environment might be in part responsible for how ɑSyn filaments assemble into phenotype-specific strains. We postulate that the maturation of ɑSyn strains develops as a function of their in vivo transmission routes and cell-specific risk factors. The impact of the cellular environment on the structural diversity of ɑSyn might have important implications for the design of preclinical studies and their use for the development of ɑSyn-based biomarkers and therapeutic strategies. By combining phenotype-specific fibrils and relevant synucleinopathy transmission models, preclinical models might more closely reflect unique disease phenotypes.
Collapse
Affiliation(s)
- Wouter Peelaerts
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
69
|
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
|
70
|
Caligiore D, Giocondo F, Silvetti M. The Neurodegenerative Elderly Syndrome (NES) hypothesis: Alzheimer and Parkinson are two faces of the same disease. IBRO Neurosci Rep 2022; 13:330-343. [PMID: 36247524 PMCID: PMC9554826 DOI: 10.1016/j.ibneur.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) and Parkinson's disease (PD) share monoamine and alpha-synuclein (αSyn) dysfunctions, often beginning years before clinical manifestations onset. The triggers for these impairments and the causes leading these early neurodegenerative processes to become AD or PD remain unclear. We address these issues by proposing a radically new perspective to frame AD and PD: they are different manifestations of one only disease we call "Neurodegenerative Elderly Syndrome (NES)". NES goes through three phases. The seeding stage, which starts years before clinical signs, and where the part of the brain-body affected by the initial αSyn and monoamine dysfunctions, influences the future possible progression of NES towards PD or AD. The compensatory stage, where the clinical symptoms are still silent thanks to compensatory mechanisms keeping monoamine concentrations homeostasis. The bifurcation stage, where NES becomes AD or PD. We present recent literature supporting NES and discuss how this hypothesis could radically change the comprehension of AD and PD comorbidities and the design of novel system-level diagnostic and therapeutic actions.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Via Sebino 32, Rome 00199, Italy
| | - Flora Giocondo
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council (LENAI-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| | - Massimo Silvetti
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| |
Collapse
|
71
|
Dadgar-Kiani E, Bieri G, Melki R, Gitler AD, Lee JH. Mesoscale connections and gene expression empower whole-brain modeling of α-synuclein spread, aggregation, and decay dynamics. Cell Rep 2022; 41:111631. [PMID: 36351406 PMCID: PMC10840492 DOI: 10.1016/j.celrep.2022.111631] [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: 08/04/2022] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022] Open
Abstract
An emerging view regarding neurodegenerative diseases is that discreet seeding of misfolded proteins leads to widespread pathology. However, the mechanisms by which misfolded proteins seed distinct brain regions and cause differential whole-brain pathology remain elusive. We used whole-brain tissue clearing and high-resolution imaging to longitudinally map pathology in an α-synuclein pre-formed fibril injection model of Parkinson's disease. Cleared brains at different time points of disease progression were quantitatively segmented and registered to a standardized atlas, revealing distinct phases of spreading and decline. We then fit a computational model with parameters that represent α-synuclein pathology spreading, aggregation, decay, and gene expression pattern to this longitudinal dataset. Remarkably, our model can generalize to predicting α-synuclein spreading patterns from several distinct brain regions and can even estimate their origins. This model empowers mechanistic understanding and accurate prediction of disease progression, paving the way for the development and testing of therapeutic interventions.
Collapse
Affiliation(s)
- Ehsan Dadgar-Kiani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Gregor Bieri
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-Aux-Roses, France
| | - Aaron D Gitler
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
72
|
Lee JH, Liu Q, Dadgar-Kiani E. Solving brain circuit function and dysfunction with computational modeling and optogenetic fMRI. Science 2022; 378:493-499. [PMID: 36327349 PMCID: PMC10543742 DOI: 10.1126/science.abq3868] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Can we construct a model of brain function that enables an understanding of whole-brain circuit mechanisms underlying neurological disease and use it to predict the outcome of therapeutic interventions? How are pathologies in neurological disease, some of which are observed to have spatial spreading mechanisms, associated with circuits and brain function? In this review, we discuss approaches that have been used to date and future directions that can be explored to answer these questions. By combining optogenetic functional magnetic resonance imaging (fMRI) with computational modeling, cell type-specific, large-scale brain circuit function and dysfunction are beginning to be quantitatively parameterized. We envision that these developments will pave the path for future therapeutics developments based on a systems engineering approach aimed at directly restoring brain function.
Collapse
Affiliation(s)
- Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, CA 94305, USA
| | - Qin Liu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Ehsan Dadgar-Kiani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
73
|
Henríquez G, Méndez L, Castañeda E, Wagler A, Jeon S, Narayan M. Preclinical Model to Evaluate Outcomes of Amyloid Cross-Toxicity in the Rodent Brain. ACS Chem Neurosci 2022; 13:2962-2973. [PMID: 36194532 DOI: 10.1021/acschemneuro.2c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The progress of neurodegenerative disorders correlates with the spread of their associated amyloidogenic proteins. Here, we investigated whether amyloid entry into nonconstitutive neurons could drive cross-toxic outcomes. Amyloid β (Aβ) was stereotaxically introduced into the rodent midbrain tegmentum, where it is not endogenously expressed. Postinfusion, rodent motor and sensorimotor capacities were assessed by standard behavioral tests at 3, 6, 9, and 12 months. The longitudinal study revealed no behavioral abnormalities. However, Aβ insult provoked intraneuronal inclusions positive for phosphorylated α-synuclein in dopaminergic neurons and were seen throughout the midbrain, a pathognomonic biomarker suggesting Parkinson's pathogenesis. These findings not only underscore the cross-toxic potential of amyloid proteins but also provide a mechanism by which they disrupt homeostasis in nonconstitutive neurons and cause neuronal corruption, injury, and demise. This study may help reconcile the large incidence of neurodegenerative comorbidity observed clinically.
Collapse
Affiliation(s)
- Gabriela Henríquez
- Department of Environmental Science and Engineering, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Lois Méndez
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Edward Castañeda
- Department of Psychology, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Amy Wagler
- Department of Mathematical Sciences, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Soyoung Jeon
- Department of Economics, Applied Statistics and International Business, New Mexico State University, Las Cruces, New Mexico 88003, United States
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| |
Collapse
|
74
|
Nayeri Z, Aliakbari F, Afzali F, Parsafar S, Gharib E, Otzen DE, Morshedi D. Characterization of exogenous αSN response genes and their relation to Parkinson’s disease using network analyses. Front Pharmacol 2022; 13:966760. [PMID: 36249814 PMCID: PMC9563388 DOI: 10.3389/fphar.2022.966760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Despite extensive research, the molecular mechanisms underlying the toxicity of αSN in Parkinson’s disease (PD) pathology are still poorly understood. To address this, we used a microarray dataset to identify genes that are induced and differentially expressed after exposure to toxic αSN aggregates, which we call exogenous αSN response (EASR) genes. Using systems biology approaches, we then determined, at multiple levels of analysis, how these EASR genes could be related to PD pathology. A key result was the identification of functional connections between EASR genes and previously identified PD-related genes by employing the proteins’ interactions networks and 9 brain region-specific co-expression networks. In each brain region, co-expression modules of EASR genes were enriched for gene sets whose expression are altered by SARS-CoV-2 infection, leading to the hypothesis that EASR co-expression genes may explain the observed links between COVID-19 and PD. An examination of the expression pattern of EASR genes in different non-neurological healthy brain regions revealed that regions with lower mean expression of the upregulated EASR genes, such as substantia nigra, are more vulnerable to αSN aggregates and lose their neurological functions during PD progression. Gene Set Enrichment Analysis of healthy and PD samples from substantia nigra revealed that a specific co-expression network, “TNF-α signaling via NF-κB”, is an upregulated pathway associated with the PD phenotype. Inhibitors of the “TNF-α signaling via NF-κB” pathway may, therefore, decrease the activity level of this pathway and thereby provide therapeutic benefits for PD patients. We virtually screened FDA-approved drugs against these upregulated genes (NR4A1, DUSP1, and FOS) using docking-based drug discovery and identified several promising drugs. Altogether, our study provides a better understanding of αSN toxicity mechanisms in PD and identifies potential therapeutic targets and small molecules for treatment of PD.
Collapse
Affiliation(s)
- Zahra Nayeri
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farhang Aliakbari
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Molecular Medicine Research Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Farzaneh Afzali
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Soha Parsafar
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ehsan Gharib
- Department of Chemistry and Biochemistry, University de Moncton, Moncton, ON, Canada
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dina Morshedi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- *Correspondence: Dina Morshedi,
| |
Collapse
|
75
|
Basaia S, Agosta F, Francia A, Cividini C, Balestrino R, Stojkovic T, Stankovic I, Markovic V, Sarasso E, Gardoni A, De Micco R, Albano L, Stefanova E, Kostic VS, Filippi M. Cerebro-cerebellar motor networks in clinical subtypes of Parkinson's disease. NPJ Parkinsons Dis 2022; 8:113. [PMID: 36068246 PMCID: PMC9448730 DOI: 10.1038/s41531-022-00377-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Parkinson's disease (PD) patients can be classified in tremor-dominant (TD) and postural-instability-and-gait-disorder (PIGD) motor subtypes. PIGD represents a more aggressive form of the disease that TD patients have a potentiality of converting into. This study investigated functional alterations within the cerebro-cerebellar system in PD-TD and PD-PIGD patients using stepwise functional connectivity (SFC) analysis and identified neuroimaging features that predict TD to PIGD conversion. Thirty-two PD-TD, 26 PD-PIGD patients and 60 healthy controls performed clinical/cognitive evaluations and resting-state functional MRI (fMRI). Four-year clinical follow-up data were available for 28 PD-TD patients, who were classified in 10 converters (cTD-PD) and 18 non-converters (ncTD-PD) to PIGD. The cerebellar seed-region was identified using a fMRI motor task. SFC analysis, characterizing regions that connect brain areas to the cerebellar seed at different levels of link-step distances, evaluated similar and divergent alterations in PD-TD and PD-PIGD. The discriminatory power of clinical data and/or SFC in distinguishing cPD-TD from ncPD-TD patients was assessed using ROC curve analysis. Compared to PD-TD, PD-PIGD patients showed decreased SFC in temporal lobe and occipital lobes and increased SFC in cerebellar cortex and ponto-medullary junction. Considering the subtype-conversion analysis, cPD-TD patients were characterized by increased SFC in temporal and occipital lobes and in cerebellum and ponto-medullary junction relative to ncPD-TD group. Combining clinical and SFC data, ROC curves provided the highest classification power to identify conversion to PIGD. These findings provide novel insights into the pathophysiology underlying different PD motor phenotypes and a potential tool for early characterization of PD-TD patients at risk of conversion to PIGD.
Collapse
Affiliation(s)
- Silvia Basaia
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Francia
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Cividini
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Roberta Balestrino
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Tanja Stojkovic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Iva Stankovic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladana Markovic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Elisabetta Sarasso
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Movement Analysis, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Gardoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Movement Analysis, San Raffaele Scientific Institute, Milan, Italy
| | - Rosita De Micco
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Luigi Albano
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elka Stefanova
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir S Kostic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
76
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
77
|
Leveraging the preformed fibril model to distinguish between alpha-synuclein inclusion- and nigrostriatal degeneration-associated immunogenicity. Neurobiol Dis 2022; 171:105804. [PMID: 35764290 PMCID: PMC9803935 DOI: 10.1016/j.nbd.2022.105804] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 01/03/2023] Open
Abstract
Neuroinflammation has become a well-accepted pathologic hallmark of Parkinson's disease (PD). However, it remains unclear whether inflammation, triggered by α-syn aggregation and/or degeneration, contributes to the progression of the disease. Studies examining neuroinflammation in PD are unable to distinguish between Lewy body-associated inflammation and degeneration-associated inflammation, as both pathologies are present simultaneously. Intrastriatal and intranigral injections of alpha-synuclein (α-syn) preformed fibrils (PFFs) results in two distinct pathologic phases: Phase 1: The accumulation and peak formation of α-syn inclusions in nigrostriatal system and, Phase 2: Protracted dopaminergic neuron degeneration. In this review we summarize the current understanding of neuroinflammation in the α-syn PFF model, leveraging the distinct Phase 1 aggregation phase and Phase 2 degeneration phase to guide our interpretations. Studies consistently demonstrate an association between pathologic α-syn aggregation in the substantia nigra (SN) and activation of the innate immune system. Further, major histocompatibility complex-II (MHC-II) antigen presentation is proportionate to inclusion load. The α-syn aggregation phase is also associated with peripheral and adaptive immune cell infiltration to the SN. These findings suggest that α-syn like aggregates are immunogenic and thus have the potential to contribute to the degenerative process. Studies examining neuroinflammation during the neurodegenerative phase reveal elevated innate, adaptive, and peripheral immune cell markers, however limitations of single time point experimental design hinder interpretations as to whether this neuroinflammation preceded, or was triggered by, nigral degeneration. Longitudinal studies across both the aggregation and degeneration phases of the model suggest that microglial activation (MHC-II) is greater in magnitude during the aggregation phase that precedes degeneration. Overall, the consistency between neuroinflammatory markers in the parkinsonian brain and in the α-syn PFF model, combined with the distinct aggregation and degenerative phases, establishes the utility of this model platform to yield insights into pathologic events that contribute to neuroinflammation and disease progression in PD.
Collapse
|
78
|
Alpha-Synuclein: The Spark That Flames Dopaminergic Neurons, In Vitro and In Vivo Evidence. Int J Mol Sci 2022; 23:ijms23179864. [PMID: 36077253 PMCID: PMC9456396 DOI: 10.3390/ijms23179864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria, α-syn fibrils and the endo-lysosomal system are key players in the pathophysiology of Parkinson’s disease. The toxicity of α-syn is amplified by cell-to-cell transmission and aggregation of endogenous species in newly invaded neurons. Toxicity of α-syn PFF was investigated using primary cultures of dopaminergic neurons or on aged mice after infusion in the SNpc and combined with mild inhibition of GBA. In primary dopaminergic neurons, application of α-syn PFF induced a progressive cytotoxicity associated with mitochondrial dysfunction, oxidative stress, and accumulation of lysosomes suggesting that exogenous α-syn reached the lysosome (from the endosome). Counteracting the α-syn endocytosis with a clathrin inhibitor, dopaminergic neuron degeneration was prevented. In vivo, α-syn PFF induced progressive neurodegeneration of dopaminergic neurons associated with motor deficits. Histology revealed progressive aggregation of α-syn and microglial activation and accounted for the seeding role of α-syn, injection of which acted as a spark suggesting a triggering of cell-to-cell toxicity. We showed for the first time that a localized SNpc α-syn administration combined with a slight lysosomal deficiency and aging triggered a progressive lesion. The cellular and animal models described could help in the understanding of the human disease and might contribute to the development of new therapies.
Collapse
|
79
|
Kukharsky MS, Everett MW, Lytkina OA, Raspopova MA, Kovrazhkina EA, Ovchinnikov RK, Antohin AI, Moskovtsev AA. Protein Homeostasis Dysregulation in Pathogenesis of Neurodegenerative Diseases. Mol Biol 2022. [DOI: 10.1134/s0026893322060115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
80
|
Hansen JY, Shafiei G, Vogel JW, Smart K, Bearden CE, Hoogman M, Franke B, van Rooij D, Buitelaar J, McDonald CR, Sisodiya SM, Schmaal L, Veltman DJ, van den Heuvel OA, Stein DJ, van Erp TGM, Ching CRK, Andreassen OA, Hajek T, Opel N, Modinos G, Aleman A, van der Werf Y, Jahanshad N, Thomopoulos SI, Thompson PM, Carson RE, Dagher A, Misic B. Local molecular and global connectomic contributions to cross-disorder cortical abnormalities. Nat Commun 2022; 13:4682. [PMID: 35948562 PMCID: PMC9365855 DOI: 10.1038/s41467-022-32420-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/28/2022] [Indexed: 12/21/2022] Open
Abstract
Numerous brain disorders demonstrate structural brain abnormalities, which are thought to arise from molecular perturbations or connectome miswiring. The unique and shared contributions of these molecular and connectomic vulnerabilities to brain disorders remain unknown, and has yet to be studied in a single multi-disorder framework. Using MRI morphometry from the ENIGMA consortium, we construct maps of cortical abnormalities for thirteen neurodevelopmental, neurological, and psychiatric disorders from N = 21,000 participants and N = 26,000 controls, collected using a harmonised processing protocol. We systematically compare cortical maps to multiple micro-architectural measures, including gene expression, neurotransmitter density, metabolism, and myelination (molecular vulnerability), as well as global connectomic measures including number of connections, centrality, and connection diversity (connectomic vulnerability). We find a relationship between molecular vulnerability and white-matter architecture that drives cortical disorder profiles. Local attributes, particularly neurotransmitter receptor profiles, constitute the best predictors of both disorder-specific cortical morphology and cross-disorder similarity. Finally, we find that cross-disorder abnormalities are consistently subtended by a small subset of network epicentres in bilateral sensory-motor, inferior temporal lobe, precuneus, and superior parietal cortex. Collectively, our results highlight how local molecular attributes and global connectivity jointly shape cross-disorder cortical abnormalities.
Collapse
Affiliation(s)
- Justine Y Hansen
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Golia Shafiei
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Jacob W Vogel
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Smart
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Martine Hoogman
- Departments of Psychiatry and Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Barbara Franke
- Departments of Psychiatry and Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Daan van Rooij
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Jan Buitelaar
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Carrie R McDonald
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Odile A van den Heuvel
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Anatomy & Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Dept of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, & Center for the Neurobiology of Leaning and Memory, University of California Irvine, 309 Qureshey Research Lab, Irvine, CA, USA
| | - Christopher R K Ching
- Keck School of Medicine, Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tomas Hajek
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Nils Opel
- Institute of Translational Psychiatry, University of Münster, Münster, Germany & Department of Psychiatry, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Gemma Modinos
- Department of Psychosis Studies & MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - André Aleman
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, Groningen, The Netherlands
| | - Ysbrand van der Werf
- Department of Anatomy & Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Neda Jahanshad
- Keck School of Medicine, Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Sophia I Thomopoulos
- Keck School of Medicine, Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Paul M Thompson
- Keck School of Medicine, Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Alain Dagher
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada.
| |
Collapse
|
81
|
Panicker N, Kam TI, Wang H, Neifert S, Chou SC, Kumar M, Brahmachari S, Jhaldiyal A, Hinkle JT, Akkentli F, Mao X, Xu E, Karuppagounder SS, Hsu ET, Kang SU, Pletnikova O, Troncoso J, Dawson VL, Dawson TM. Neuronal NLRP3 is a parkin substrate that drives neurodegeneration in Parkinson's disease. Neuron 2022; 110:2422-2437.e9. [PMID: 35654037 PMCID: PMC9357148 DOI: 10.1016/j.neuron.2022.05.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 04/09/2022] [Accepted: 05/12/2022] [Indexed: 02/09/2023]
Abstract
Parkinson's disease (PD) is mediated, in part, by intraneuronal accumulation of α-synuclein aggregates andsubsequent death of dopamine (DA) neurons in the substantia nigra pars compacta (SNpc). Microglial hyperactivation of the NOD-like receptor protein 3 (NLRP3) inflammasome has been well-documented in various neurodegenerative diseases, including PD. We show here that loss of parkin activity in mouse and human DA neurons results in spontaneous neuronal NLRP3 inflammasome assembly, leading to DA neuron death. Parkin normally inhibits inflammasome priming by ubiquitinating and targeting NLRP3 for proteasomal degradation. Loss of parkin activity also contributes to the assembly of an active NLRP3 inflammasome complex via mitochondrial-derived reactive oxygen species (mitoROS) generation through the accumulation of another parkin ubiquitination substrate, ZNF746/PARIS. Inhibition of neuronal NLRP3 inflammasome assembly prevents degeneration of DA neurons in familial and sporadic PD models. Strategies aimed at limiting neuronal NLRP3 inflammasome activation hold promise as a disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Nikhil Panicker
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
| | - Hu Wang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Stewart Neifert
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Shih-Ching Chou
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Manoj Kumar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Saurav Brahmachari
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aanishaa Jhaldiyal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jared T Hinkle
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fatih Akkentli
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Enquan Xu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Eric T Hsu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juan Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA.
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA.
| |
Collapse
|
82
|
Neuronal ApoE Regulates the Cell-to-Cell Transmission of α-Synuclein. Int J Mol Sci 2022; 23:ijms23158311. [PMID: 35955451 PMCID: PMC9369063 DOI: 10.3390/ijms23158311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
The presence of protein inclusions, called Lewy bodies (LBs) and Lewy neurites (LNs), in the brain is the main feature of Parkinson’s disease (PD). Recent evidence that the prion-like propagation of α-synuclein (α-syn), as a major component of LBs and LNs, plays an important role in the progression of PD has gained much attention, although the molecular mechanism remains unclear. In this study, we evaluated whether neuronal ApoE regulates the cell-to-cell transmission of α-syn and explored its molecular mechanism using in vitro and in vivo model systems. We demonstrate that neuronal ApoE deficiency attenuates both α-syn uptake and release by downregulating LRP-1 and LDLR expression and enhancing chaperone-mediated autophagy activity, respectively, thereby contributing to α-syn propagation. In addition, we observed that α-syn propagation was attenuated in ApoE knockout mice injected with pre-formed mouse α-syn fibrils. This study will help our understanding of the molecular mechanisms underlying α-syn propagation.
Collapse
|
83
|
Revell AY, Silva AB, Arnold TC, Stein JM, Das SR, Shinohara RT, Bassett DS, Litt B, Davis KA. A framework For brain atlases: Lessons from seizure dynamics. Neuroimage 2022; 254:118986. [PMID: 35339683 PMCID: PMC9342687 DOI: 10.1016/j.neuroimage.2022.118986] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/13/2022] [Accepted: 02/07/2022] [Indexed: 01/03/2023] Open
Abstract
Brain maps, or atlases, are essential tools for studying brain function and organization. The abundance of available atlases used across the neuroscience literature, however, creates an implicit challenge that may alter the hypotheses and predictions we make about neurological function and pathophysiology. Here, we demonstrate how parcellation scale, shape, anatomical coverage, and other atlas features may impact our prediction of the brain's function from its underlying structure. We show how network topology, structure-function correlation (SFC), and the power to test specific hypotheses about epilepsy pathophysiology may change as a result of atlas choice and atlas features. Through the lens of our disease system, we propose a general framework and algorithm for atlas selection. This framework aims to maximize the descriptive, explanatory, and predictive validity of an atlas. Broadly, our framework strives to provide empirical guidance to neuroscience research utilizing the various atlases published over the last century.
Collapse
Affiliation(s)
- Andrew Y Revell
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Alexander B Silva
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of California, San Francisco, CA 94143, USA
| | - T Campbell Arnold
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joel M Stein
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandhitsu R Das
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Statistics in Imaging and Visualization Endeavor, Perelman school of Medicine, University of Pennsylvania, PA 19104, USA
| | - Dani S Bassett
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Department of Electrical and Systems Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physics and Astronomy, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Brian Litt
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn A Davis
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
84
|
Chen L, Nagaraja C, Daniels S, Fisk ZA, Dvorak R, Meyerdirk L, Steiner JA, Escobar Galvis ML, Henderson MX, Rousseaux MWC, Brundin P, Chu HY. Synaptic location is a determinant of the detrimental effects of α-Synuclein pathology to glutamatergic transmission in the basolateral amygdala. eLife 2022; 11:78055. [PMID: 35775627 PMCID: PMC9286736 DOI: 10.7554/elife.78055] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
The presynaptic protein α-synuclein (αSyn) has been suggested to be involved in the pathogenesis of Parkinson’s disease (PD). In PD, the amygdala is prone to develop insoluble αSyn aggregates, and it has been suggested that circuit dysfunction involving the amygdala contributes to the psychiatric symptoms. Yet, how αSyn aggregates affect amygdala function is unknown. In this study, we examined αSyn in glutamatergic axon terminals and the impact of its aggregation on glutamatergic transmission in the basolateral amygdala (BLA). We found that αSyn is primarily present in the vesicular glutamate transporter 1-expressing (vGluT1+) terminals in the mouse BLA, which is consistent with higher levels of αSyn expression in vGluT1+ glutamatergic neurons in the cerebral cortex relative to the vGluT2+ glutamatergic neurons in the thalamus. We found that αSyn aggregation selectively decreased the cortico-BLA, but not the thalamo-BLA, transmission; and that cortico-BLA synapses displayed enhanced short-term depression upon repetitive stimulation. In addition, using confocal microscopy, we found that vGluT1+ axon terminals exhibited decreased levels of soluble αSyn, which suggests that lower levels of soluble αSyn might underlie the enhanced short-term depression of cortico-BLA synapses. In agreement with this idea, we found that cortico-BLA synaptic depression was also enhanced in αSyn knockout mice. In conclusion, both basal and dynamic cortico-BLA transmission were disrupted by abnormal aggregation of αSyn and these changes might be relevant to the perturbed cortical control of the amygdala that has been suggested to play a role in psychiatric symptoms in PD.
Collapse
Affiliation(s)
- Liqiang Chen
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | - Chetan Nagaraja
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | - Samuel Daniels
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | - Zoe A Fisk
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Rachel Dvorak
- Department of Neurodegenerative Science, Van Andel Institute, GRand Rapids, United States
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | - Jennifer A Steiner
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | | | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| | - Maxime W C Rousseaux
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Patrik Brundin
- Pharma Research and Early Development (pRED), F. Hoffmann-La Roche, Little Falls, United States
| | - Hong-Yuan Chu
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, United States
| |
Collapse
|
85
|
Tsafaras G, Baekelandt V. The role of LRRK2 in the periphery: link with Parkinson's disease and inflammatory diseases. Neurobiol Dis 2022; 172:105806. [PMID: 35781002 DOI: 10.1016/j.nbd.2022.105806] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/30/2022] [Accepted: 06/22/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is currently considered a multisystemic disorder rather than a pure brain disease, in line with the multiple hit hypothesis from Braak. However, despite increasing evidence that the pathology might originate in the periphery, multiple unknown aspects and contradictory data on the pathological processes taking place in the periphery jeopardize the interpretation and therapeutic targeting of PD. Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene have been widely linked with familial and sporadic PD cases. However, the actual role of LRRK2 in PD pathophysiology is far from understood. There is evidence that LRRK2 may be involved in alpha-synuclein (α-synuclein) pathology and immune cell regulation, but it has also been associated with inflammatory diseases such as inflammatory bowel disease, tuberculosis, leprosy, and several other bacterial infections. In this review, we focus on the different roles of LRRK2 in the periphery. More specifically, we discuss the involvement of LRRK2 in the propagation of α-synuclein pathology and its regulatory role in peripheral inflammation. A deeper understanding of the multidimensional functions of LRRK2 will pave the way for more accurate characterization of PD pathophysiology and its association with other inflammatory diseases.
Collapse
Affiliation(s)
- George Tsafaras
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
86
|
Quantum-dot-labeled synuclein seed assay identifies drugs modulating the experimental prion-like transmission. Commun Biol 2022; 5:636. [PMID: 35768587 PMCID: PMC9243017 DOI: 10.1038/s42003-022-03590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/15/2022] [Indexed: 11/08/2022] Open
Abstract
Synucleinopathies are neurodegenerative disorders including Parkinson disease (PD), dementia with Lewy body (DLB), and multiple system atrophy (MSA) that involve deposits of the protein alpha-synuclein (α-syn) in the brain. The inoculation of α-syn aggregates derived from synucleinopathy or preformed fibrils (PFF) formed in vitro induces misfolding and deposition of endogenous α-syn. This is referred to as prion-like transmission, and the mechanism is still unknown. In this study, we label α-syn PFF with quantum dots and visualize their movement directly in acute slices of brain tissue inoculated with α-syn PFF seeds. Using this system, we find that the trafficking of α-syn seeds is dependent on fast axonal transport and the seed spreading is dependent on endocytosis and neuronal activity. We also observe pharmacological effects on α-syn seed spreading; clinically available drugs including riluzole are effective in reducing the spread of α-syn seeds and this effect is also observed in vivo. Our quantum-dot-labeled α-syn seed assay system combined with in vivo transmission experiment reveals an early phase of transmission, in which uptake and spreading of seeds occur depending on neuronal activity, and a later phase, in which seeds induce the propagation of endogenous misfolded α-syn.
Collapse
|
87
|
Kulkarni AS, Burns MR, Brundin P, Wesson DW. Linking α-synuclein-induced synaptopathy and neural network dysfunction in early Parkinson's disease. Brain Commun 2022; 4:fcac165. [PMID: 35822101 PMCID: PMC9272065 DOI: 10.1093/braincomms/fcac165] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/11/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
The prodromal phase of Parkinson's disease is characterized by aggregation of the misfolded pathogenic protein α-synuclein in select neural centres, co-occurring with non-motor symptoms including sensory and cognitive loss, and emotional disturbances. It is unclear whether neuronal loss is significant during the prodrome. Underlying these symptoms are synaptic impairments and aberrant neural network activity. However, the relationships between synaptic defects and network-level perturbations are not established. In experimental models, pathological α-synuclein not only impacts neurotransmission at the synaptic level, but also leads to changes in brain network-level oscillatory dynamics-both of which likely contribute to non-motor deficits observed in Parkinson's disease. Here we draw upon research from both human subjects and experimental models to propose a 'synapse to network prodrome cascade' wherein before overt cell death, pathological α-synuclein induces synaptic loss and contributes to aberrant network activity, which then gives rise to prodromal symptomology. As the disease progresses, abnormal patterns of neural activity ultimately lead to neuronal loss and clinical progression of disease. Finally, we outline goals and research needed to unravel the basis of functional impairments in Parkinson's disease and other α-synucleinopathies.
Collapse
Affiliation(s)
- Aishwarya S Kulkarni
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Matthew R Burns
- Department of Neurology, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Patrik Brundin
- Pharma Research and Early Development (pRED), F. Hoffman-La Roche, Little Falls, NJ, USA
| | - Daniel W Wesson
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| |
Collapse
|
88
|
Henderson MX, Henrich MT, Geibl FF, Oertel WH, Brundin P, Surmeier DJ. The roles of connectivity and neuronal phenotype in determining the pattern of α-synuclein pathology in Parkinson's disease. Neurobiol Dis 2022; 168:105687. [PMID: 35283326 PMCID: PMC9610381 DOI: 10.1016/j.nbd.2022.105687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, and motor dysfunction has been attributed to loss of dopaminergic neurons. However, motor dysfunction is only one of many symptoms experienced by patients. A neuropathological hallmark of PD is intraneuronal protein aggregates called Lewy pathology (LP). Neuropathological staging studies have shown that dopaminergic neurons are only one of the many cell types prone to manifest LP. Progressive appearance of LP in multiple brain regions, as well as peripheral nerves, has led to the popular hypothesis that LP and misfolded forms of one of its major components - α-synuclein (aSYN) - can spread through synaptically connected circuits. However, not all brain regions or neurons within connected circuits develop LP, suggesting that cell autonomous factors modulate the development of pathology. Here, we review studies about how LP develops and progressively engages additional brain regions. We focus on how connectivity constrains progression and discuss cell autonomous factors that drive pathology development. We propose a mixed model of cell autonomous factors and trans-synaptic spread as mediators of pathology progression and put forward this model as a framework for future experiments exploring PD pathophysiology.
Collapse
Affiliation(s)
- Michael X Henderson
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| | - Martin T Henrich
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Fanni F Geibl
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany
| | - Patrik Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| |
Collapse
|
89
|
Initiation and progression of α-synuclein pathology in Parkinson’s disease. Cell Mol Life Sci 2022; 79:210. [PMID: 35347432 PMCID: PMC8960654 DOI: 10.1007/s00018-022-04240-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/21/2022]
Abstract
α-Synuclein aggregation is a critical molecular process that underpins the pathogenesis of Parkinson’s disease. Aggregates may originate at synaptic terminals as a consequence of aberrant interactions between α-synuclein and lipids or evasion of proteostatic defences. The nature of these interactions is likely to influence the emergence of conformers or strains that in turn could explain the clinical heterogeneity of Parkinson’s disease and related α-synucleinopathies. For neurodegeneration to occur, α-synuclein assemblies need to exhibit seeding competency, i.e. ability to template further aggregation, and toxicity which is at least partly mediated by interference with synaptic vesicle or organelle homeostasis. Given the dynamic and reversible conformational plasticity of α-synuclein, it is possible that seeding competency and cellular toxicity are mediated by assemblies of different structure or size along this continuum. It is currently unknown which α-synuclein assemblies are the most relevant to the human condition but recent advances in the cryo-electron microscopic characterisation of brain-derived fibrils and their assessment in stem cell derived and animal models are likely to facilitate the development of precision therapies or biomarkers. This review summarises the main principles of α-synuclein aggregate initiation and propagation in model systems, and their relevance to clinical translation.
Collapse
|
90
|
Lopes DM, Llewellyn SK, Harrison IF. Propagation of tau and α-synuclein in the brain: therapeutic potential of the glymphatic system. Transl Neurodegener 2022; 11:19. [PMID: 35314000 PMCID: PMC8935752 DOI: 10.1186/s40035-022-00293-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Many neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease, are characterised by the accumulation of misfolded protein deposits in the brain, leading to a progressive destabilisation of the neuronal network and neuronal death. Among the proteins that can abnormally accumulate are tau and α-synuclein, which can propagate in a prion-like manner and which upon aggregation, represent the most common intracellular proteinaceous lesions associated with neurodegeneration. For years it was thought that these intracellular proteins and their accumulation had no immediate relationship with extracellular homeostasis pathways such as the glymphatic clearance system; however, mounting evidence has now suggested that this is not the case. The involvement of the glymphatic system in neurodegenerative disease is yet to be fully defined; however, it is becoming increasingly clear that this pathway contributes to parenchymal solute clearance. Importantly, recent data show that proteins prone to intracellular accumulation are subject to glymphatic clearance, suggesting that this system plays a key role in many neurological disorders. In this review, we provide a background on the biology of tau and α-synuclein and discuss the latest findings on the cell-to-cell propagation mechanisms of these proteins. Importantly, we discuss recent data demonstrating that manipulation of the glymphatic system may have the potential to alleviate and reduce pathogenic accumulation of propagation-prone intracellular cytotoxic proteins. Furthermore, we will allude to the latest potential therapeutic opportunities targeting the glymphatic system that might have an impact as disease modifiers in neurodegenerative diseases.
Collapse
|
91
|
Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell Mol Life Sci 2022; 79:174. [PMID: 35244787 PMCID: PMC8897347 DOI: 10.1007/s00018-022-04166-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 12/18/2022]
Abstract
Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer’s disease or Parkinson’s disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the β-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.
Collapse
|
92
|
Extracellular alpha-synuclein: Sensors, receptors, and responses. Neurobiol Dis 2022; 168:105696. [DOI: 10.1016/j.nbd.2022.105696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 11/19/2022] Open
|
93
|
Yoshida S, Hasegawa T. Deciphering the prion-like behavior of pathogenic protein aggregates in neurodegenerative diseases. Neurochem Int 2022; 155:105307. [PMID: 35181393 DOI: 10.1016/j.neuint.2022.105307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are hitherto classified based on their core clinical features, the anatomical distribution of neurodegeneration, and the cell populations mainly affected. On the other hand, the wealth of neuropathological, genetic, molecular and biochemical studies have identified the existence of distinct insoluble protein aggregates in the affected brain regions. These findings have spread the use of a collective term, proteinopathy, for neurodegenerative disorders with particular type of structurally altered protein accumulation. Particularly, a recent breakthrough in this field came with the discovery that these protein aggregates can transfer from one cell to another, thereby converting normal proteins to potentially toxic, misfolded species in a prion-like manner. In this review, we focus specifically on the molecular and cellular basis that underlies the seeding activity and transcellular spreading phenomenon of neurodegeneration-related protein aggregates, and discuss how these events contribute to the disease progression.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan; Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Yamagata, 992-1202, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan.
| |
Collapse
|
94
|
Chen H, Wang K, Scheperjans F, Killinger B. Environmental triggers of Parkinson's disease - Implications of the Braak and dual-hit hypotheses. Neurobiol Dis 2022; 163:105601. [PMID: 34954321 PMCID: PMC9525101 DOI: 10.1016/j.nbd.2021.105601] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 11/22/2022] Open
Abstract
Idiopathic Parkinson's disease (PD) may take decades to develop, during which many risk or protective factors may come into play to initiate the pathogenesis or modify its progression to clinical PD. The lack of understanding of this prodromal phase of PD and the factors involved has been a major hurdle in the study of PD etiology and preventive strategies. Although still controversial, the Braak and dual-hit hypotheses that PD may start peripherally in the olfactory structures and/or the gut provides a theoretical platform to identify the triggers and modifiers of PD prodromal development and progression. This is particularly true for the search of environmental causes of PD as the olfactory structures and gut are the major human mucosal interfaces with the environment. In this review, we lay out our personal views about how the Braak and dual-hit hypotheses may help us search for the environmental triggers and modifiers for PD, summarize available experimental and epidemiological evidence, and discuss research gaps and strategies.
Collapse
Affiliation(s)
- Honglei Chen
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Keran Wang
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital, and Clinicum, University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Bryan Killinger
- Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
95
|
Van Den Berge N, Ulusoy A. Animal models of brain-first and body-first Parkinson's disease. Neurobiol Dis 2022; 163:105599. [DOI: 10.1016/j.nbd.2021.105599] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
|
96
|
Zhou D, Lynn CW, Cui Z, Ciric R, Baum GL, Moore TM, Roalf DR, Detre JA, Gur RC, Gur RE, Satterthwaite TD, Bassett DS. Efficient coding in the economics of human brain connectomics. Netw Neurosci 2022; 6:234-274. [PMID: 36605887 PMCID: PMC9810280 DOI: 10.1162/netn_a_00223] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
In systems neuroscience, most models posit that brain regions communicate information under constraints of efficiency. Yet, evidence for efficient communication in structural brain networks characterized by hierarchical organization and highly connected hubs remains sparse. The principle of efficient coding proposes that the brain transmits maximal information in a metabolically economical or compressed form to improve future behavior. To determine how structural connectivity supports efficient coding, we develop a theory specifying minimum rates of message transmission between brain regions to achieve an expected fidelity, and we test five predictions from the theory based on random walk communication dynamics. In doing so, we introduce the metric of compression efficiency, which quantifies the trade-off between lossy compression and transmission fidelity in structural networks. In a large sample of youth (n = 1,042; age 8-23 years), we analyze structural networks derived from diffusion-weighted imaging and metabolic expenditure operationalized using cerebral blood flow. We show that structural networks strike compression efficiency trade-offs consistent with theoretical predictions. We find that compression efficiency prioritizes fidelity with development, heightens when metabolic resources and myelination guide communication, explains advantages of hierarchical organization, links higher input fidelity to disproportionate areal expansion, and shows that hubs integrate information by lossy compression. Lastly, compression efficiency is predictive of behavior-beyond the conventional network efficiency metric-for cognitive domains including executive function, memory, complex reasoning, and social cognition. Our findings elucidate how macroscale connectivity supports efficient coding and serve to foreground communication processes that utilize random walk dynamics constrained by network connectivity.
Collapse
Affiliation(s)
- Dale Zhou
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher W. Lynn
- Initiative for the Theoretical Sciences, Graduate Center, City University of New York, New York, NY, USA,Joseph Henry Laboratories of Physics, Princeton University, Princeton, NJ, USA
| | - Zaixu Cui
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rastko Ciric
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, CA, USA
| | - Graham L. Baum
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Tyler M. Moore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, Philadelphia, PA, USA
| | - David R. Roalf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A. Detre
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben C. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, Philadelphia, PA, USA
| | - Raquel E. Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, Philadelphia, PA, USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, Philadelphia, PA, USA
| | - Dani S. Bassett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Physics & Astronomy, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA,Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA,Department of Electrical & Systems Engineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA,Santa Fe Institute, Santa Fe, NM, USA,* Corresponding Author:
| |
Collapse
|
97
|
Awa S, Suzuki G, Masuda-Suzukake M, Nonaka T, Saito M, Hasegawa M. Phosphorylation of endogenous α-synuclein induced by extracellular seeds initiates at the pre-synaptic region and spreads to the cell body. Sci Rep 2022; 12:1163. [PMID: 35064139 PMCID: PMC8782830 DOI: 10.1038/s41598-022-04780-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of phosphorylated α-synuclein aggregates has been implicated in several diseases, such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB), and is thought to spread in a prion-like manner. Elucidating the mechanisms of prion-like transmission of α-synuclein is important for the development of therapies for these diseases, but little is known about the details. Here, we injected α-synuclein fibrils into the brains of wild-type mice and examined the early phase of the induction of phosphorylated α-synuclein accumulation. We found that phosphorylated α-synuclein appeared within a few days after the intracerebral injection. It was observed initially in presynaptic regions and subsequently extended its localization to axons and cell bodies. These results suggest that extracellular α-synuclein fibrils are taken up into the presynaptic region and seed-dependently convert the endogenous normal α-synuclein that is abundant there to an abnormal phosphorylated form, which is then transported through the axon to the cell body.
Collapse
Affiliation(s)
- Shiori Awa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Genjiro Suzuki
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Masami Masuda-Suzukake
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Nonaka
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Minoru Saito
- Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan.,Department of Correlative Study in Physics and Chemistry, Graduate School of Integrated Basic Sciences, Nihon University, Tokyo, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
98
|
Stress-inducible phosphoprotein 1 (HOP/STI1/STIP1) regulates the accumulation and toxicity of α-synuclein in vivo. Acta Neuropathol 2022; 144:881-910. [PMID: 36121476 PMCID: PMC9547791 DOI: 10.1007/s00401-022-02491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/26/2023]
Abstract
The predominantly pre-synaptic intrinsically disordered protein α-synuclein is prone to misfolding and aggregation in synucleinopathies, such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). Molecular chaperones play important roles in protein misfolding diseases and members of the chaperone machinery are often deposited in Lewy bodies. Here, we show that the Hsp90 co-chaperone STI1 co-immunoprecipitated α-synuclein, and co-deposited with Hsp90 and Hsp70 in insoluble protein fractions in two mouse models of α-synuclein misfolding. STI1 and Hsp90 also co-localized extensively with filamentous S129 phosphorylated α-synuclein in ubiquitin-positive inclusions. In PD human brains, STI1 transcripts were increased, and in neurologically healthy brains, STI1 and α-synuclein transcripts correlated. Nuclear Magnetic Resonance (NMR) analyses revealed direct interaction of α-synuclein with STI1 and indicated that the STI1 TPR2A, but not TPR1 or TPR2B domains, interacted with the C-terminal domain of α-synuclein. In vitro, the STI1 TPR2A domain facilitated S129 phosphorylation by Polo-like kinase 3. Moreover, mice over-expressing STI1 and Hsp90ß presented elevated α-synuclein S129 phosphorylation accompanied by inclusions when injected with α-synuclein pre-formed fibrils. In contrast, reduced STI1 function decreased protein inclusion formation, S129 α-synuclein phosphorylation, while mitigating motor and cognitive deficits as well as mesoscopic brain atrophy in α-synuclein-over-expressing mice. Our findings reveal a vicious cycle in which STI1 facilitates the generation and accumulation of toxic α-synuclein conformers, while α-synuclein-induced proteostatic stress increased insoluble STI1 and Hsp90.
Collapse
|
99
|
Rahayel S, Mišić B, Zheng YQ, Liu ZQ, Abdelgawad A, Abbasi N, Caputo A, Zhang B, Lo A, Kehm V, Kozak M, Soo Yoo H, Dagher A, Luk KC. Differentially targeted seeding reveals unique pathological alpha-synuclein propagation patterns. Brain 2021; 145:1743-1756. [PMID: 34910119 PMCID: PMC9166565 DOI: 10.1093/brain/awab440] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/21/2021] [Accepted: 11/04/2021] [Indexed: 11/14/2022] Open
Abstract
Parkinson's Disease is a progressive neurodegenerative disorder characterized by the intracellular accumulation of insoluble alpha-synuclein aggregates into Lewy bodies and neurites. Increasing evidence indicates that Parkinson's Disease progression results from the spread of pathologic alpha-synuclein through neuronal networks. However, the exact mechanisms underlying the propagation of abnormal proteins in the brain are only partially understood. The objective of this study was first to describe the long-term spatiotemporal distributions of Lewy-related pathology in mice injected with alpha-synuclein preformed fibrils and then to recreate these patterns using a computational model that simulates in silico the spread of pathologic alpha-synuclein. In this study, 87 two-to-three-month-old non-transgenic mice were injected with alpha-synuclein preformed fibrils to generate a comprehensive post-mortem dataset representing the long-term spatiotemporal distributions of hyperphosphorylated alpha-synuclein, an established marker of Lewy pathology, across the 426 regions of the Allen Mouse Brain Atlas. The mice were injected into either the caudoputamen, nucleus accumbens or hippocampus and followed over 24 months with pathologic alpha-synuclein quantified at seven intermediate time points. The pathologic patterns observed at each time point in this high-resolution dataset were then compared to those generated using a Susceptible-Infected-Removed computational model, an agent-based model that simulates the spread of pathologic alpha-synuclein for every brain region taking simultaneously into account the effect of regional brain connectivity and Snca gene expression. Our histopathological findings showed that differentially targeted seeding of pathologic alpha-synuclein resulted in unique propagation patterns over 24 months and that most brain regions were permissive to pathology. We found that the Susceptible-Infected-Removed model recreated the observed distributions of pathology over 24 months for each injection site. Null models showed that both Snca gene expression and connectivity had a significant influence on model fit. In sum, our study demonstrates that the combination of normal alpha-synuclein concentration and brain connectomics contributes to making brain regions more vulnerable to the pathological process, providing support for a prion-like spread of pathologic alpha-synuclein. We propose that this rich dataset and the related computational model will help test new hypotheses regarding mechanisms that may alter the spread of pathologic alpha-synuclein in the brain.
Collapse
Affiliation(s)
- Shady Rahayel
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada.,Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec H4J 1C5, Canada
| | - Bratislav Mišić
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Ying-Qiu Zheng
- Wellcome Centre for Integrative Neuroimaging, Centre for Functional Magnetic Resonance Imaging of the Brain, University of Oxford, John Radcliffe Hospital, Oxford, Oxfordshire, UK
| | - Zhen-Qi Liu
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Alaa Abdelgawad
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Nooshin Abbasi
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Anna Caputo
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| | - Bin Zhang
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| | - Angela Lo
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| | - Victoria Kehm
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| | - Michael Kozak
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| | - Han Soo Yoo
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA.,Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Alain Dagher
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-4283, USA
| |
Collapse
|
100
|
Zhang Q, Abdelmotilib H, Larson T, Keomanivong C, Conlon M, Aldridge GM, Narayanan NS. Cortical alpha-synuclein preformed fibrils do not affect interval timing in mice. Neurosci Lett 2021; 765:136273. [PMID: 34601038 DOI: 10.1016/j.neulet.2021.136273] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
One hallmark feature of Parkinson's disease (PD) is Lewy body pathology associated with misfolded alpha-synuclein. Previous studies have shown that striatal injection of alpha-synuclein preformed fibrils (PFF) can induce misfolding and aggregation of native alpha-synuclein in a prion-like manner, leading to cell death and motor dysfunction in mouse models. Here, we tested whether alpha-synuclein PFFs injected into the medial prefrontal cortex results in deficits in interval timing, a cognitive task which is disrupted in human PD patients and in rodent models of PD. We injected PFF or monomers of human alpha-synuclein into the medial prefrontal cortex of mice pre-injected with adeno-associated virus (AAV) coding for overexpression of human alpha-synuclein or control protein. Despite notable medial prefrontal cortical synucleinopathy, we did not observe consistent deficits in fixed-interval timing. These results suggest that cortical alpha-synuclein does not reliably disrupt fixed-interval timing.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States.
| | - Hisham Abdelmotilib
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Travis Larson
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Cameron Keomanivong
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Mackenzie Conlon
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Georgina M Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | | |
Collapse
|