1
|
Kallunki P, Sotty F, Willén K, Lubas M, David L, Ambjørn M, Bergström AL, Buur L, Malik I, Nyegaard S, Eriksen TT, Krogh BO, Stavenhagen JB, Andersen KJ, Pedersen LØ, Cholak E, van den Brink EN, Rademaker R, Vink T, Satijn D, Parren PWHI, Christensen S, Olsen LR, Søderberg JN, Vergo S, Jensen A, Egebjerg J, Wulff-Larsen PG, Harndahl MN, Damlund DSM, Bjerregaard-Andersen K, Fog K. Rational selection of the monoclonal α-synuclein antibody amlenetug (Lu AF82422) for the treatment of α-synucleinopathies. NPJ Parkinsons Dis 2025; 11:132. [PMID: 40404755 PMCID: PMC12098740 DOI: 10.1038/s41531-024-00849-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/28/2024] [Indexed: 05/24/2025] Open
Abstract
Amlenetug (Lu AF82422) is a human monoclonal antibody targeting α-synuclein in clinical development for multiple system atrophy. We describe a series of studies that characterize its functional properties and supported its selection as a viable clinical candidate. Amlenetug inhibits seeding induced in mouse primary neurons by various α-synuclein fibrillar assemblies and by aggregates isolated from MSA brain homogenate. In vivo, both co-injection of amlenetug with α-synuclein assemblies in mouse brain and peripheral administration inhibit α-synuclein seeding. Amlenetug inhibits uptake of α-synuclein seeds as well as accumulation of C-terminal truncated α-synuclein seeds and demonstrates binding to monomeric, aggregated, and truncated forms of human α-synuclein. The epitope of amlenetug was mapped to amino acids 112-117 and further characterized by crystallographic structure analysis. Based on our data, we hypothesize that targeting α-synuclein will potentially slow further disease progression by inhibiting further pathology development but be without impact on established pathology and symptoms.
Collapse
Affiliation(s)
- Pekka Kallunki
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark.
| | - Florence Sotty
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Katarina Willén
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Michal Lubas
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Laurent David
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Malene Ambjørn
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | - Louise Buur
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Ibrahim Malik
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | | | - Berit O Krogh
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | | | - Lars Ø Pedersen
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Ersoy Cholak
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | - Rik Rademaker
- Genmab, Uppsalalaan 15, 3584 CT, Utrecht, The Netherlands
| | - Tom Vink
- Genmab, Uppsalalaan 15, 3584 CT, Utrecht, The Netherlands
| | - David Satijn
- Genmab, Uppsalalaan 15, 3584 CT, Utrecht, The Netherlands
| | | | | | - Line R Olsen
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | - Sandra Vergo
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Allan Jensen
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | - Jan Egebjerg
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| | | | | | | | | | - Karina Fog
- H. Lundbeck A/S, Research, Ottiliavej 9, 2500, Valby, Denmark
| |
Collapse
|
2
|
Al-Lahham R, Corkins ME, Ishtikhar M, Rabadia P, Ramirez S, Banerjee V, Shahnawaz M. Intracellular Inclusions Induced by Patient-Derived and Amplified α-Synuclein Aggregates Are Morphologically Indistinguishable. Cells 2025; 14:684. [PMID: 40422187 PMCID: PMC12110328 DOI: 10.3390/cells14100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
Lewy Body Disease (LBD) and Multiple System Atrophy (MSA) are synucleinopathies with distinct prognoses and neuropathologies, however, with overlapping clinical symptoms. Different disease characteristics are proposed to be determined by distinct conformations of alpha-synuclein (α-Syn) aggregates, which can self-propagate and spread between cells via a prion-like mechanism. The goal of this study is to investigate whether α-syn aggregates amplified from brain and CSF samples of LBD and MSA patients using the Seed Amplification Assay (SAA) maintain α-Syn seeding properties similar to those of α-syn aggregates derived from patients' brains. To address this, SAA-amplified and un-amplified α-Syn aggregates from LBD and MSA patients' brains, as well as SAA-amplified α-Syn aggregates from LBD and MSA patients' CSF samples, were used to treat synuclein biosensor cells, and induced intracellular α-Syn inclusions were analyzed by confocal microscopy. Our data indicate that induced α-Syn aggregates from LBD and MSA patients' brains have similar seeding properties and morphological characteristics in the α-Syn biosensor cells as those amplified from LBD and MSA patients' brains, as well as those amplified from LBD and MSA patients' CSF samples. In this study, we demonstrated that, regardless of the source of aggregates, the seeds from LBD and MSA produce cellular accumulation of α-Syn with distinct morphologies, confirming the presence of different conformational strains of α-Syn in LBD and MSA and allowing us to differentiate synucleinopathies based on the morphology of aggregates and seeding properties.
Collapse
Affiliation(s)
- Rabab Al-Lahham
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| | - Mark E. Corkins
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Mohd Ishtikhar
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| | - Prakruti Rabadia
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| | - Santiago Ramirez
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| | - Victor Banerjee
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| | - Mohammad Shahnawaz
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.I.); (P.R.); (S.R.)
| |
Collapse
|
3
|
Uemura N. Fibril-seeded animal models of synucleinopathies: Pathological mechanisms, disease modeling, and therapeutic implications. Neurosci Res 2025:S0168-0102(25)00082-3. [PMID: 40316176 DOI: 10.1016/j.neures.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Accumulating evidence suggests that prion-like spread of misfolded α-Synuclein (αSyn) underlies the pathological progression of Lewy body diseases (LBD). Animal models injected with αSyn preformed fibrils (PFFs) have provided strong evidence for the prion hypothesis in LBD. Moreover, αSyn PFFs can be administered to various hosts and regions, contributing to the elucidation of pathological mechanisms and disease modeling. These models have also been used to identify biomarkers and develop new disease-modifying therapies for LBD. In contrast, it remains unknown how the prion-like properties of αSyn contribute to the pathogenesis of multiple system atrophy (MSA). Recent studies indicate that conformationally distinct αSyn fibrils induce different pathological features in animals, supporting the strain hypothesis, which suggests that conformational variations in αSyn fibrils contribute to the clinicopathological heterogeneity in synucleinopathies. However, the study of disease-specific αSyn fibrils in pathological mechanisms and disease modeling is still in its early stages. This review aims to highlight recent advances in αSyn fibril-seeded animal models with an emphasis on their unique features and utility in exploring pathological mechanisms and identifying novel disease-modifying therapies. In addition, I discuss future directions for refining these models in light of the emerging strain hypothesis in synucleinopathies.
Collapse
Affiliation(s)
- Norihito Uemura
- Department of Neurological Disease Control, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; Department of Therapeutics for Multiple System Atrophy, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyoku, Kyoto 606-8507, Japan.
| |
Collapse
|
4
|
Zacharopoulou M, Seetaloo N, Ross J, Stephens AD, Fusco G, McCoy TM, Dai W, Mela I, Fernandez-Villegas A, Martel A, Routh AF, De Simone A, Phillips JJ, Kaminski Schierle GS. Local Ionic Conditions Modulate the Aggregation Propensity and Influence the Structural Polymorphism of α-Synuclein. J Am Chem Soc 2025; 147:13131-13145. [PMID: 40207671 PMCID: PMC12023029 DOI: 10.1021/jacs.4c13473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
Parkinson's disease (PD) is linked to the aggregation of the intrinsically disordered protein α-synuclein (aSyn), but the precise triggers and mechanisms driving this process remain unclear. Local environmental factors, such as ion concentrations, can influence aSyn's conformational ensemble and its tendency to aggregate. In this study, we explore how physiologically relevant ions, mainly Ca2+ and Na+, affect aSyn aggregation, monomer structural dynamics, and fibril polymorphism. ThT fluorescence assays show that all ions speed up aggregation, with Ca2+ having the strongest effect. Using heteronuclear single quantum correlation nuclear magnetic resonance (1H-15N HSQC NMR) spectroscopy, we validate that Ca2+ binds at the C-terminus while Na+ interacts nonspecifically across the sequence. Small-angle neutron scattering (SANS) and hydrogen-deuterium exchange mass spectrometry (HDX-MS) show that Na+ leads to more extended aSyn structures, while Ca2+ results in moderate extension. Molecular dynamics (MD) simulations support this, showing Na+ increases extension between the NAC region and C-terminus, whereas Ca2+ biases the ensemble toward a moderately elongated structure. MD also shows that Ca2+ increases water persistence times in the hydration shell, indicating that aSyn aggregation propensity is due to a combination of conformational bias of the monomer and solvent mobility. Atomic force microscopy (AFM) points toward the formation of distinct fibril polymorphs under different ionic conditions, suggesting ion-induced monomer changes contribute to the diversity of fibril structures. These findings underscore the pivotal influence of the local ionic milieu in shaping the structure and aggregation propensity of aSyn, offering insights into the molecular underpinnings of PD and potential therapeutic strategies targeting aSyn dynamics.
Collapse
Affiliation(s)
- Maria Zacharopoulou
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Neeleema Seetaloo
- Living
Systems Institute, University of Exeter, Stocker Road, Exeter EX4 4QD, U.K.
| | - James Ross
- School
of Molecular and Cellular Biology and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K.
| | - Amberley D. Stephens
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Giuliana Fusco
- Department
of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
- Department
of Life Sciences, Imperial College London, London SW7 2AZ, U.K.
| | - Thomas M. McCoy
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Wenyue Dai
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Ioanna Mela
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Ana Fernandez-Villegas
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Anne Martel
- Institut
Laue Langevin, 71 Avenue
des Martyrs, Grenoble CS 20156 38042, France
| | - Alexander F. Routh
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Alfonso De Simone
- Department
of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
- Department
of Life Sciences, Imperial College London, London SW7 2AZ, U.K.
| | - Jonathan J. Phillips
- Living
Systems Institute, University of Exeter, Stocker Road, Exeter EX4 4QD, U.K.
| | - Gabriele S. Kaminski Schierle
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| |
Collapse
|
5
|
Raina A, Wang W, Gonzalez JC, Yan X, Overstreet-Wadiche L, Wadiche JI, Zhang CL, Chen SG. Distinct alpha-synuclein strains derived from Parkinson's disease patient tissues trigger differential inclusion pathology in a novel biosensor cell model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646513. [PMID: 40236210 PMCID: PMC11996501 DOI: 10.1101/2025.04.01.646513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background α-Synuclein (αSyn) can misfold and aggregate to form fibrillar ß-sheet-rich aggregates ("strains") that are phosphorylated (p-αSyn) and deposited into intracellular inclusions in the brain, the pathological hallmark of synucleinopathies including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Previously, we reported that seed amplification assays such as real-time quaking-induced conversion (RT-QuIC) amplifies and detects αSyn strains from the patient skin. However, whether skin-derived αSyn strains induce disease-specific pathological features in a biological system is unknown. Methods We generated a U251 human glioblastoma cell line expressing fluorescently tagged αSyn carrying the PD-linked A53T mutation and Förster resonance energy transfer (FRET)-based U251 biosensor cells. Using fluorescence microscopy coupled with in situ detergent extraction, FRET-Flow cytometry and high-content confocal imaging, we examined the pathological burden and morphology of p-αSyn inclusions seeded by RT-QuIC-amplified patient skin and brain αSyn strains in αSyn-expressing U251 cells, FRET-based αSyn biosensor cells and αSyn biosensor cell-derived neurons. Results U251 cells allow robust and rapid in situ detection of detergent-insoluble intracellular αSyn inclusions triggered by exogenous αSyn seeds. In U251 FRET-based biosensor cells, PD skin-amplified strains induce a greater pathological burden and distinct p-αSyn inclusion morphology from PD brain-amplified and DLB skin-amplified strains. Inclusion morphology of DLB and MSA skin- and brain-amplified strains are comparable. Furthermore, skin-amplified αSyn strains induce neuronal inclusions and cause degeneration of induced neurons reprogrammed from the U251 biosensor cells. Finally, biosensor cell-propagated PD skin αSyn strains induce higher seeding activity measured by RT-QuIC than PD brain and DLB skin αSyn strains, linking intracellular pathological burden to in vitro seeding activity. Conclusions We report the detection of distinct PD strains derived from patient skin and brain tissues that trigger unique pathological phenotypes in U251 αSyn biosensor cells and cause degeneration of reprogrammed neurons from the same cell lineage. Moreover, DLB and MSA skin αSyn strains mimic pathological features of their brain αSyn strains in these biosensor cells. Therefore, the U251 αSyn biosensor cell model is a robust tool to potentially discriminate PD and DLB synucleinopathies and to study αSyn tissue- and strain-specific etiology and pathogenesis. Graphical abstract
Collapse
|
6
|
Jellinger KA. Pathomechanisms of neuropsychiatric disturbances in atypical parkinsonian disorders: a current view. J Neural Transm (Vienna) 2025; 132:495-518. [PMID: 39954076 DOI: 10.1007/s00702-025-02890-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025]
Abstract
Multiple system atrophy (MSA), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) are the most common atypical parkinsonisms. These adult-onset and lethal neurodegenerative disorders of unknown etiology are clinically characterized by varying combinations of autonomic, levodopa-poorly responsive parkinsonsm, motor, non-motor, cerebellar syndromes, behavioral, cognitive and other neuropsychiatric disorders. Although their pathological hallmarks are different-MSA α-synucleinopathy, CBD and PSP 4-repeat (4R) tauopathies-their neuropsychiatric disturbances include anxiety, depression, agitations, attention-executive dysfunctions, less often compulsive and REM sleep behavior disorders (RBD), which may contribute to disease progression and reduced quality of life (QoL) of patients and caregivers. The present paper reviews the prevalence and type of neuropsychiatric profile in these atypical parkinsonian syndromes, their neuroimaging, and pathogenic backgrounds based on extensive literature research. MSA patients show anxiety, apathy (depression), initial RBD, attentional and executive dysfunction; PSP patients present with apathy, depression, disinhibition, and to a lesser extent, anxiety and agitation; CBD patients are featured by executive and visuospatial dysfunctions, irritability, alien limb phenomena, sleep and language disorders. Neuropsychiatric disorders in these syndromes are often similar, due to disruption of prefronto-subcortical (limbic) and striato-thalamo-cortical circuitries or default mode and attention network disorder. This supports the concept that they are brain network disorders due to complex pathogenic mechanisms related to the basic proteinopathies that are still poorly understood. Psychotic symptoms, hallucinations and delusions are rare. Neuropsychiatric changes in these disorders are often premature and anticipate motor dysfunctions; their assessment and further elucidation of their pathogenesis are warranted as a basis for early diagnosis and adequate treatment of these debilitating comorbidities.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
7
|
Koss DJ, Todd O, Menon H, Anderson Z, Yang T, Findlay L, Graham B, Palmowski P, Porter A, Morrice N, Walker L, Attems J, Ghanem SS, El-Agnaf O, LeBeau FE, Erskine D, Outeiro TF. A reciprocal relationship between markers of genomic DNA damage and alpha-synuclein pathology in dementia with Lewy bodies. Mol Neurodegener 2025; 20:34. [PMID: 40114198 PMCID: PMC11927131 DOI: 10.1186/s13024-025-00813-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND DNA damage and DNA damage repair (DDR) dysfunction are insults with broad implications for cellular physiology and have been implicated in various neurodegenerative diseases. Alpha-synuclein (aSyn), a pre-synaptic and nuclear protein associated with neurodegenerative disorders known as synucleinopathies, has been associated with DNA double strand break (DSB) repair. However, although nuclear aSyn pathology has been observed in cortical tissue of dementia with Lewy body (DLB) cases, whether such nuclear pathology coincides with the occurrence of DNA damage has not previously been investigated. Moreover, the specific types of DNA damage elevated in DLB cases and the contribution of DNA damage towards Lewy body (LB) formation is unknown. METHODS DNA damage and aSyn pathology were assessed in fixed lateral temporal cortex from clinically and neuropathologically confirmed DLB cases and controls, as well as in cortical tissue from young 3-month-old presymptomatic A30P-aSyn mice. Frozen lateral temporal cortex from DLB and control cases was subject to nuclear isolation, western blotting, aSyn seed amplification and proteomic characterisation via mass spectrometry. RESULTS We detected seed-competent nuclear aSyn, and elevated nuclear serine-129 phosphorylation in DLB temporal cortex, alongside the accumulation of DSBs in neuronal and non-neuronal cellular populations. DNA damage was also present in cortical tissue from presymptomatic A30P mice, demonstrating it is an early insult closely associated with pathogenic aSyn. Strikingly, in postmortem DLB tissue, markers of genomic DNA damage-derived cytoplasmic DNA (CytoDNA) were evident within the majority of LBs examined. The observed cellular pathology was consistent with nuclear upregulation of associated DDR proteins, particularly those involved in base excision repair and DSB repair pathways. CONCLUSIONS Collectively our study demonstrates the accumulation of seed-competent pathological nuclear associated aSyn, alongside nuclear DNA damage and the potential involvement of DNA damage derived cytoDNA species in cytoplasmic aSyn pathology. Ultimately, our study supports the hypothesis of a reciprocal relationship between aSyn pathology and nuclear DNA damage and highlights a potential underlying role for DNA damage in pathological mechanisms relevant to DLB, as well as other synucleinopathies, opening novel possibilities for diagnosis and treatment.
Collapse
Affiliation(s)
- David J Koss
- Division of Neuroscience, School of Medicine, University of Dundee, Dundee, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK.
| | - Olivia Todd
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Hariharan Menon
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Zoe Anderson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Tamsin Yang
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Lucas Findlay
- Division of Neuroscience, School of Medicine, University of Dundee, Dundee, UK
| | - Ben Graham
- Division of Neuroscience, School of Medicine, University of Dundee, Dundee, UK
| | - Pawel Palmowski
- Newcastle University Protein and Proteome Analysis Unit, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew Porter
- Newcastle University Protein and Proteome Analysis Unit, Newcastle University, Newcastle Upon Tyne, UK
| | - Nicola Morrice
- Division of Neuroscience, School of Medicine, University of Dundee, Dundee, UK
| | - Lauren Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Simona S Ghanem
- Neurological Disorders Research Centre, Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Omar El-Agnaf
- Neurological Disorders Research Centre, Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Fiona En LeBeau
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle, UK
| | - Daniel Erskine
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Tiago F Outeiro
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK.
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Scientific Employee With an Honorary Contract at Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.
| |
Collapse
|
8
|
Masters CL. Neuropathology meets chemical and genetic pathology head-on: a personal perspective. Pathology 2025; 57:191-195. [PMID: 39668075 DOI: 10.1016/j.pathol.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 12/14/2024]
Abstract
Medical science has made revolutionary discoveries around the nosology and aetiology of the neurodegenerative diseases. Dementia is the second leading cause of death in Australia. My colleagues and I are now looking at therapeutics which potentially can delay or prevent the onset of Alzheimer's disease (AD). Advances in diagnosis allow detection of preclinical AD. Neuropathologists working closely with chemical and genetic pathologists have a major role to play in pushing diagnostics and therapeutics to the forefront of clinical management of these diseases.
Collapse
Affiliation(s)
- Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Vic, Australia.
| |
Collapse
|
9
|
Liu H, Yu R, Zhang M, Zheng X, Zhong L, Yang W, Luo Y, Huang Z, Zheng J, Zhong H, Wei X, Zheng W, Yu Y, Wang Q. Fibrinogen degradation products exacerbate alpha-synuclein aggregation by inhibiting autophagy via downregulation of Beclin1 in multiple system atrophy. Neurotherapeutics 2025; 22:e00538. [PMID: 39904669 PMCID: PMC12014411 DOI: 10.1016/j.neurot.2025.e00538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/24/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disease arising from accumulation of the α-synuclein and aberrant protein clearance in oligodendrocytes. The mechanisms of autophagy involved in the progression of MSA remain poorly understood. It is reported that MSA patients have blood-brain barrier impairments, which may increase the entry of fibrinogen into the brain. However, the roles of fibrinogen and its degradation products (FDPs) on autophagy and α-synuclein accumulation in MSA remain unknown. Here, we established the MSA animal model by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) and 3-nitropropionic acid (3-NP), and cellular models by adding fibrillar α-syn into oligodendrocytes to investigate the mechanisms of FDPs on autophagy and accumulation of α-synuclein in oligodendrocytes. We found that FDPs inhibit the entry of α-synuclein into lysosomes for degradation, increasing aggregation of α-synuclein in oligodendrocytes (OLN-93). Our findings indicated that in OLN-93, FDPs inhibited the expressions of Beclin1 and Bif-1, which could promote the fusion of autophagosomes with lysosomes. Furthermore, the expression of α-synuclein was elevated in FDPs-injected mice, accompanied by an increase in the protein level of p62. We detected elevated expression of FDPs in the striatum of MSA mice. Finally, FDPs inhibited the expression of Beclin1 and Bif-1, which led to aberrant autophagic degradation and increased aggregation of α-synuclein and phospho-α-synuclein in MSA mice. Our study illustrates that FDPs can cause aggregation of α-synuclein in MSA by inhibiting Beclin1-mediated autophagy, which may exacerbate disease progression. These results provide a new therapeutic approach for MSA, that targets the inhibitory effect of FDPs on oligodendrocyte autophagy.
Collapse
Affiliation(s)
- Huanzhu Liu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Ruoyang Yu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Muwei Zhang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Lizi Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hui Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wenhua Zheng
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, 999078, Macao, China
| | - Yinghua Yu
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, Australia.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
10
|
Heras-Garvin A, Fellner L, Granata R, Wenning GK, Stefanova N. Transcriptional dysregulation in the cerebellum triggered by oligodendroglial α-synucleinopathy: insights from a transgenic mouse into the early disease mechanisms of MSA. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02892-5. [PMID: 39954078 DOI: 10.1007/s00702-025-02892-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disorder characterized by abnormal accumulation of α-synuclein, progressive neuronal loss, motor impairment and widespread pathological changes, which include significant involvement of the cerebellum. To understand the early molecular mechanisms that might underlie α-synuclein-triggered MSA cerebellar pathology, we performed RNA sequencing (RNA-Seq) of cerebellar samples from a well-established model of MSA. RNA-Seq and differential gene expression analysis was conducted in the PLP-αSyn model of MSA. Cerebellum from two and 12-month-old MSA and wildtype mice were used. Gene ontology (GO) and KEGG enrichment analyses of the differentially expressed genes (DEGs) were performed to explore processes involved in MSA-like disease progression. The overlap between transcriptional changes in MSA and those associated with aging was also evaluated. RNA-Seq analysis demonstrated significant transcriptional dysregulation in cerebellum from MSA mice, even at early stages. GO and KEGG analyses of DEGs point to a potential role of synaptic dysfunction, cellular signaling dysregulation and inflammation in the cerebellar pathology of MSA mice. In addition, those changes exacerbate with disease progression. Additionally, our analysis of aging in both control and PLP-αSyn mice showed that age-related transcriptional changes in mid-aged controls seem to be present in young MSA mice. Thus, MSA-like pathology might lead to an acceleration of aging-related mechanisms. Our findings demonstrate significant cerebellar transcriptional dysregulation triggered by oligodendroglial α-synucleinopathy in PLP-αSyn mice, revealing pathways that might be critical for the early cerebellar pathology of MSA, and that may serve as potential molecular targets for therapeutic interventions in this devastating disorder.
Collapse
Affiliation(s)
- Antonio Heras-Garvin
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innrain 66, 3rd floor, Innsbruck, 6020, Austria.
| | - Lisa Fellner
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innrain 66, 3rd floor, Innsbruck, 6020, Austria
| | - Roberta Granata
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innrain 66, 3rd floor, Innsbruck, 6020, Austria
| | - Gregor K Wenning
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innrain 66, 3rd floor, Innsbruck, 6020, Austria
| | - Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innrain 66, 3rd floor, Innsbruck, 6020, Austria.
| |
Collapse
|
11
|
McGregor BA, Raihan MO, Brishti A, Hur J, Porter JE. Deciphering motor dysfunction and microglial activation in mThy1- α-synuclein mice: a comprehensive study of behavioral, gene expression, and methylation changes. Front Mol Neurosci 2025; 18:1544971. [PMID: 40018011 PMCID: PMC11865073 DOI: 10.3389/fnmol.2025.1544971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Growing recognition of microglia's role in neurodegenerative disorders has accentuated the need to characterize microglia profiles and their influence on pathogenesis. To understand changes observed in the microglial profile during the progression of synucleinopathies, microglial gene expression and DNA methylation were examined in the mThy1-α-synuclein mouse model. Methods Disease progression was determined using behavioral tests evaluating locomotor deficits before DNA and RNA extraction at 7 and 10 months from isolated microglia for enzymatic methyl-sequencing and RNA-sequencing. Results Pathway analysis of these changes at 7 months indicates a pro-inflammatory profile and changes in terms related to synaptic maintenance. Expression and methylation at both 7 and 10 months included terms regarding mitochondrial and metabolic stress. While behavior symptoms progressed at 10 months, we see many previously activated pathways being inhibited in microglia at a later stage, with only 8 of 53 shared pathways predicted to be directionally concordant. Despite the difference in pathway directionality, 21 of the 22 genes that were differentially expressed and annotated to differentially methylated regions at both 7 and 10 months had conserved directionality changes. Discussion These results highlight a critical period in disease progression, during which the microglia respond to α-synuclein, suggesting a transition in the role of microglia from the early to late stages of the disease.
Collapse
Affiliation(s)
- Brett A. McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Md. Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
- Department of Pharmaceutical Sciences, Chicago State University School of Pharmacy, Chicago, IL, United States
| | - Afrina Brishti
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James E. Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
12
|
Wiseman JA, Turner CP, Faull RLM, Halliday GM, Dieriks BV. Refining α-synuclein seed amplification assays to distinguish Parkinson's disease from multiple system atrophy. Transl Neurodegener 2025; 14:7. [PMID: 39920796 PMCID: PMC11804046 DOI: 10.1186/s40035-025-00469-6] [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: 08/01/2024] [Accepted: 01/14/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) and multiple system atrophy (MSA) are two distinct α-synucleinopathies traditionally differentiated through clinical symptoms. Early diagnosis of MSA is problematic, and seed amplification assays (SAAs), such as real-time quaking-induced conversion (RT-QuIC), offer the potential to distinguish these diseases through their underlying α-synuclein (α-Syn) pathology and proteoforms. Currently, SAAs provide a binary result, signifying either the presence or absence of α-Syn seeds. To enhance the diagnostic potential and biological relevance of these assays, there is a pressing need to incorporate quantification and stratification of α-Syn proteoform-specific aggregation kinetics into current SAA pipelines. METHODS Optimal RT-QuIC assay conditions for α-Syn seeds extracted from PD and MSA patient brains were determined, and assay kinetics were assessed for α-Syn seeds from different pathologically relevant brain regions (medulla, substantia nigra, hippocampus, middle temporal gyrus, and cerebellum). The conformational profiles of disease- and region-specific α-Syn proteoforms were determined by subjecting the amplified reaction products to concentration-dependent proteolytic digestion with proteinase K. RESULTS Using our protocol, PD and MSA could be accurately delineated using proteoform-specific aggregation kinetics, including α-Syn aggregation rate, maximum relative fluorescence, the gradient of amplification, and core protofilament size. MSA cases yielded significantly higher values than PD cases across all four kinetic parameters in brain tissues, with the MSA-cerebellar phenotype having higher maximum relative fluorescence than the MSA-Parkinsonian phenotype. Statistical significance was maintained when the data were analysed regionally and when all regions were grouped. CONCLUSIONS Our RT-QuIC protocol and analysis pipeline can distinguish between PD and MSA, and between MSA phenotypes. MSA α-Syn seeds induce faster propagation and exhibit higher aggregation kinetics than PD α-Syn, mirroring the biological differences observed in brain tissue. With further validation of these quantitative parameters, we propose that SAAs could advance from a yes/no diagnostic to a theranostic biomarker that could be utilised in developing therapeutics.
Collapse
Affiliation(s)
- James A Wiseman
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Clinton P Turner
- LabPlus, Department of Anatomical Pathology, Te Whatu Ora, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand
| | - Glenda M Halliday
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia
- Neuroscience Research Australia & Faculty of Medicine School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Birger Victor Dieriks
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand.
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand.
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia.
| |
Collapse
|
13
|
Bendetowicz D, Planche V, Bezard E, Dehay B, Meissner WG. Biological definitions of synucleinopathies should be anchored in clinical trajectories and encompass the complex biology of the disease. JOURNAL OF PARKINSON'S DISEASE 2025:1877718X241313443. [PMID: 39973487 DOI: 10.1177/1877718x241313443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Recently, two proposals for defining Parkinson's disease and its related pathogenic processes have been published. In this viewpoint, we discuss the primary drivers behind these efforts, the future directions, and the challenges that must be addressed. While finding biomarkers is a mandatory step for better precision medicine and optimal patient stratification in therapeutic trials, we argue that a biological definition of Parkinson's disease based on a single biomarker will struggle to account for the complexity of the mechanisms involved in developing the disease. Additionally, a biological definition of asymptomatic patients should rely on a thorough understanding of patients' clinical trajectories, which is currently not the case in synucleinopathies.
Collapse
Affiliation(s)
- David Bendetowicz
- Univ. Bordeaux, CNRS, IMN, UMR5293, Bordeaux, France
- CHU Bordeaux, Service de Neurologie des Maladies Neurodégénératives, IMNc, Centre Expert Parkinson, NS-Park/FCRIN Network, Bordeaux, France
| | - Vincent Planche
- Univ. Bordeaux, CNRS, IMN, UMR5293, Bordeaux, France
- CHU de Bordeaux, Service de Neurologie des Maladies Neurodégénératives, IMNc, Centre Mémoire de Ressources et de Recherche, Bordeaux, France
| | - Erwan Bezard
- Univ. Bordeaux, CNRS, IMN, UMR5293, Bordeaux, France
| | | | - Wassilios G Meissner
- Univ. Bordeaux, CNRS, IMN, UMR5293, Bordeaux, France
- CHU Bordeaux, Service de Neurologie des Maladies Neurodégénératives, IMNc, Centre Expert Parkinson, NS-Park/FCRIN Network, Bordeaux, France
- Department of Medicine, University of Otago and New Zealand Brain Research Institute, Christchurch, New Zealand
| |
Collapse
|
14
|
Wiseman JA, Reddy K, Dieriks BV. From onset to advancement: the temporal spectrum of α-synuclein in synucleinopathies. Ageing Res Rev 2025; 104:102640. [PMID: 39667671 DOI: 10.1016/j.arr.2024.102640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/21/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
This review provides an in-depth analysis of the complex role of alpha-synuclein (α-Syn) in the development of α-synucleinopathies, with a particular focus on its structural diversity and the resulting clinical variability. The ability of α-Syn to form different strains or polymorphs and undergo various post-translational modifications significantly contributes to the wide range of symptoms observed in disorders such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), as well as in lesser-known non-classical α-synucleinopathies. The interaction between genetic predispositions and environmental factors further complicates α-synucleinopathic disease pathogenesis, influencing the disease-specific onset and progression. Despite their common pathological hallmark of α-Syn accumulation, the clinical presentation and progression of α-synucleinopathies differ significantly, posing challenges for diagnosis and treatment. The intricacies of α-Syn pathology highlight the critical need for a deeper understanding of its biological functions and interactions within the neuronal environment to develop targeted therapeutic strategies. The precise point at which α-Syn aggregation transitions from being a byproduct of initial disease triggers to an active and independent driver of disease progression - through the propagation and acceleration of pathogenic processes - remains unclear. By examining the role of α-Syn across various contexts, we illuminate its dual role as both a marker and a mediator of disease, offering insights that could lead to innovative approaches for managing α-synucleinopathies.
Collapse
Affiliation(s)
- James A Wiseman
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand; Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kreesan Reddy
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand; Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - Birger Victor Dieriks
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand; Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia.
| |
Collapse
|
15
|
Jellinger KA. The Spectrum of Cognitive Impairment in Atypical Parkinsonism Syndromes: A Comprehensive Review of Current Understanding and Research. Diseases 2025; 13:39. [PMID: 39997046 PMCID: PMC11854393 DOI: 10.3390/diseases13020039] [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: 12/23/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) are the most common atypical parkinsonism (AP) syndromes. They are clinically characterized by varying combinations of levodopa-poorly responsive parkinsonism, motor, cerebellar, and other signs. They are associated with a wide spectrum of non-motor symptoms, including prominent cognitive impairment such as global cognitive deficits, memory, executive, attentional, visuospatial, language, and non-verbal reasoning dysfunctions. Within the APs, their cognitive functioning is distributed along a continuum from MSA with the least impaired cognitive profile (similar to Parkinson's disease) to PSP and CBD with the greatest decline in global cognitive and executive domains. Although their pathological hallmarks are different-MSA α-synucleinopathy, CBD, and PSP 4-repeat tauopathies-cognitive dysfunctions in APs show both overlaps and dissimilarities. They are often preceding and anticipate motor dysfunctions, finally contributing to reduced quality of life of patients and caregivers. The present paper will review the current evidence of the prevalence and type of cognitive impairment in these AP syndromes, their neuroimaging, pathogenic backgrounds, and current management options based on extensive literature research. Cognitive dysfunctions in APs are due to disruption of prefronto-subcortical and striato-thalamo-cortical circuitries and multiple essential brain networks. This supports the concept that they are brain network disorders due to complex pathogenic mechanisms related to the basic proteinopathies that are still poorly understood. Therefore, the pathophysiology and pathogenesis of cognitive impairment in APs deserve further elucidation as a basis for early diagnosis and adequate treatment of these debilitating comorbidities.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
16
|
Bräuer S, Schniewind I, Dinter E, Falkenburger BH. Recursive seed amplification detects distinct α-synuclein strains in cerebrospinal fluid of patients with Parkinson's disease. Acta Neuropathol Commun 2025; 13:13. [PMID: 39833972 PMCID: PMC11749544 DOI: 10.1186/s40478-024-01923-8] [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: 11/22/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder with a wide range of clinical phenotypes. Pathologically, it is characterized by neuronal inclusions containing misfolded, fibrillar alpha-synuclein (aSyn). Prion-like properties of aSyn contribute to the spread of aSyn pathology throughout the nervous system as the disease progresses. Utilizing these properties, seed amplification assays (SAA) enable the detection of aSyn pathology in living patients. We hypothesized that structurally distinct aSyn aggregates, or strains, may underlie the clinical heterogeneity of PD. To test this hypothesis, we recursively amplified aSyn fibrils from the cerebrospinal fluid (CSF) of 54 patients (34 people with PD and 20 controls). These fibrils were then characterized regarding SAA kinetic properties and detergent resistance. In addition, cultured cells were transfected with SAA products, and the extent of seeded aSyn pathology was quantified by staining for phosphorylated aSyn followed by automated high-throughput microscopy and image analysis. We found that fibrils, amplified from CSF by recursive SAA, exhibit two types of distinct biophysical properties and have different seeding capacities in cells. These properties are associated with clinical parameters and may therefore help explain the clinical heterogeneity in PD. Measuring aSyn strains may be relevant for prognosis and for therapies targeting aSyn pathology.
Collapse
Affiliation(s)
- Stefan Bräuer
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
| | - Iñaki Schniewind
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
| | - Elisabeth Dinter
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
| | - Björn H Falkenburger
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany.
| |
Collapse
|
17
|
Ndayisaba A, Halliday GM, Khurana V. Multiple System Atrophy: Pathology, Pathogenesis, and Path Forward. ANNUAL REVIEW OF PATHOLOGY 2025; 20:245-273. [PMID: 39405585 DOI: 10.1146/annurev-pathmechdis-051122-104528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease characterized by autonomic failure and motor impairment. The hallmark pathologic finding in MSA is widespread oligodendroglial cytoplasmic inclusions rich in aggregated α-synuclein (αSyn). MSA is widely held to be an oligodendroglial synucleinopathy, and we outline lines of evidence to support this assertion, including the presence of early myelin loss. However, we also consider emerging data that support the possibility of neuronal or immune dysfunction as a primary driver of MSA. These hypotheses are placed in the context of a major recent discovery that αSyn is conformationally distinct in MSA versus other synucleinopathies such as Parkinson's disease. We outline emerging techniques in epidemiology, genetics, and molecular pathology that will shed more light on this mysterious disease. We anticipate a future in which cutting-edge developments in personalized disease modeling, including with pluripotent stem cells, bridge mechanistic developments at the bench and real benefits at the bedside.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Division of Movement Disorders, Ann Romney Center for Neurologic Diseases, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | - Glenda M Halliday
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Vikram Khurana
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Division of Movement Disorders, Ann Romney Center for Neurologic Diseases, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA;
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, Maryland, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Park H, Kam TI, Dawson VL, Dawson TM. α-Synuclein pathology as a target in neurodegenerative diseases. Nat Rev Neurol 2025; 21:32-47. [PMID: 39609631 DOI: 10.1038/s41582-024-01043-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
α-Synuclein misfolds into pathological forms that lead to various neurodegenerative diseases known collectively as α-synucleinopathies. In this Review, we provide a comprehensive overview of pivotal advances in α-synuclein research. We examine structural features and physiological functions of α-synuclein and summarize current insights into key post-translational modifications, such as nitration, phosphorylation, ubiquitination, sumoylation and truncation, considering their contributions to neurodegeneration. We also highlight the existence of disease-specific α-synuclein strains and their mechanisms of pathological spread, and discuss seed amplification assays and PET tracers as emerging diagnostic tools for detecting pathological α-synuclein in clinical settings. We also discuss α-synuclein aggregation and clearance mechanisms, and review cell-autonomous and non-cell-autonomous processes that contribute to neuronal death, including the roles of adaptive and innate immunity in α-synuclein-driven neurodegeneration. Finally, we highlight promising therapeutic approaches that target pathological α-synuclein and provide insights into emerging areas of research.
Collapse
Affiliation(s)
- Hyejin Park
- Neuroregeneration and Stem Cell Programs, 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
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, 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
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, 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
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, 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.
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Wiseman JA, Fu Y, Faull RLM, Turner CP, Curtis MA, Halliday GM, Dieriks BV. N-terminus α-synuclein detection reveals new and more diverse aggregate morphologies in multiple system atrophy and Parkinson's disease. Transl Neurodegener 2024; 13:67. [PMID: 39726015 DOI: 10.1186/s40035-024-00456-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) and multiple system atrophy (MSA) are classified as α-synucleinopathies and are primarily differentiated by their clinical phenotypes. Delineating these diseases based on their specific α-synuclein (α-Syn) proteoform pathologies is crucial for accurate antemortem biomarker diagnosis. Newly identified α-Syn pathologies in PD raise questions about whether MSA exhibits a similar diversity. This prompted the need for a comparative study focusing on α-Syn epitope-specific immunoreactivities in both diseases, which could clarify the extent of pathological overlap and diversity, and guide more accurate biomarker development. METHODS We utilised a multiplex immunohistochemical approach to detect multiple structural domains of α-Syn proteoforms across multiple regions prone to pathological accumulation in MSA (n = 10) and PD (n = 10). Comparison of epitope-specific α-Syn proteoforms was performed in the MSA medulla, inferior olivary nucleus, substantia nigra, hippocampus, and cerebellum, and in the PD olfactory bulb, medulla, substantia nigra, hippocampus, and entorhinal cortex. RESULTS N-terminus and C-terminus antibodies detected significantly more α-Syn pathology in MSA than antibodies for phosphorylated (pS129) α-Syn, which are classically used to detect α-Syn. Importantly, C-terminus immunolabelling is more pronounced in MSA compared to PD. Meanwhile, N-terminus immunolabelling consistently detected the highest percentage of α-Syn across pathologically burdened regions of both diseases, which could be of biological significance. As expected, oligodendroglial involvement distinguished MSA from PD, but in contrast to PD, no substantial astrocytic or microglial α-Syn accumulation in MSA occurred. These data confirm glial-specific changes between these diseases when immunolabelling the N-terminus epitope. In comparison, N-terminus neuronal α-Syn was present in PD and MSA, with most MSA neurons lacking pS129 α-Syn proteoforms. This explains why characterisation of neuronal MSA pathologies is lacking and challenges the reliance on pS129 antibodies for the accurate quantification of α-Syn pathological load across α-synucleinopathies. CONCLUSIONS These findings underscore the necessity of utilising a multiplex approach to detect α-Syn, most importantly including the N-terminus, to capture the entire spectrum of α-Syn proteoforms in α-synucleinopathies. The data provide novel insights toward the biological differentiation of these α-synucleinopathies and pave the way for more refined antemortem diagnostic methods to facilitate early identification and intervention of these neurodegenerative diseases.
Collapse
Affiliation(s)
- James A Wiseman
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, , Auckland, 1142, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia
| | - YuHong Fu
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, , Auckland, 1142, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand
| | - Clinton P Turner
- LabPlus, Department of Anatomical Pathology, Te Whatu Ora, Auckland, New Zealand
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, , Auckland, 1142, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand
| | - Glenda M Halliday
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia
- Neuroscience Research Australia & Faculty of Medicine School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Birger V Dieriks
- Department of Anatomy and Medical Imaging, University of Auckland, 85 Park Road, Grafton, , Auckland, 1142, New Zealand.
- Centre for Brain Research, University of Auckland, Auckland, 1023, New Zealand.
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, NSW, 2050, Australia.
| |
Collapse
|
20
|
Krismer F, Fanciulli A, Meissner WG, Coon EA, Wenning GK. Multiple system atrophy: advances in pathophysiology, diagnosis, and treatment. Lancet Neurol 2024; 23:1252-1266. [PMID: 39577925 DOI: 10.1016/s1474-4422(24)00396-x] [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: 06/14/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/24/2024]
Abstract
Multiple system atrophy is an adult-onset, sporadic, and progressive neurodegenerative disease. People with this disorder report a wide range of motor and non-motor symptoms. Overlap in the clinical presentation of multiple system atrophy with other movement disorders (eg, Parkinson's disease and progressive supranuclear palsy) is a concern for accurate and timely diagnosis. Over the past 5 years, progress has been made in understanding key pathophysiological events in multiple system atrophy, including the seeding of α-synuclein inclusions and the detection of disease-specific α-synuclein strains. Diagnostic criteria were revised in 2022 with the intention to improve the accuracy of a diagnosis of multiple system atrophy, particularly for early disease stages. Early signals of efficacy in clinical trials have indicated the potential for disease-modifying therapies for multiple system atrophy, although no trial has yet provided unequivocal evidence of neuroprotection in this rare disease. The advances in pathophysiology could play a part in biomarker discovery for early diagnosis as well as in the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Florian Krismer
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria.
| | | | - Wassilios G Meissner
- Centre Hospitalier Universitaire Bordeaux, Service de Neurologie des Maladies Neurodégénératives, Institut des Maladies Neurodégénératives Clinique, French Clinical Research Network for Parkinson's Disease and Movement Disorders, Bordeaux, France; Université de Bordeaux, Centre National de la Recherche Scientifique, Institut des Maladies Neurodégénératives, Unité Mixte de Recherche 5293, Bordeaux, France; Department of Medicine, University of Otago, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | | | - Gregor K Wenning
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Niu J, Zhong Y, Xue L, Wang H, Hu D, Liao Y, Zhang X, Dou X, Yu C, Wang B, Sun Y, Tian M, Zhang H, Wang J. Spatial-temporal dynamic evolution of lewy body dementia by metabolic PET imaging. Eur J Nucl Med Mol Imaging 2024; 52:145-157. [PMID: 39155308 DOI: 10.1007/s00259-024-06881-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE Lewy body dementia (LBD) is a neurodegenerative disease with high heterogeneity and complex pathogenesis. Our study aimed to use disease progression modeling to uncover spatial-temporal dynamic evolution of LBD in vivo, and to explore differential profiles of clinical features, glucose metabolism, and dopaminergic function among different evolution-related subtypes. METHODS A total of 123 participants (31 healthy controls and 92 LBD patients) who underwent 18F-FDG PET scans were retrospectively enrolled. 18F-FDG PET-based Subtype and Stage Inference (SuStaIn) model was established to illustrate spatial-temporal evolutionary patterns and categorize relevant subtypes. Then subtypes and stages were further related to clinical features, glucose metabolism, and dopaminergic function of LBD patients. RESULTS This 18F-FDG PET imaging-based approach illustrated two distinct patterns of neurodegenerative evolution originating from the neocortex and basal ganglia in LBD and defined them as subtype 1 and subtype 2, respectively. There were obvious differences between subtypes. Compared with subtype 1, subtype 2 exhibited a greater proportion of male patients (P = 0.045) and positive symptoms such as visual hallucinations (P = 0.033) and fluctuating cognitions (P = 0.033). Cognitive impairment, metabolic abnormalities, dopaminergic dysfunction and progression were all more severe in subtype 2 (all P < 0.05). In addition, a strong association was observed between SuStaIn subtypes and two clinical phenotypes (Parkinson's disease dementia and dementia with Lewy bodies) (P = 0.005). CONCLUSIONS Our findings based on 18F-FDG PET and data-driven model illustrated spatial-temporal dynamic evolution of LBD and categorized novel subtypes with different evolutionary patterns, clinical and imaging features in vivo. The evolution-related subtypes are associated with LBD clinical phenotypes, which supports the perspective of existence of distinct entities in LBD spectrum.
Collapse
Affiliation(s)
- Jiaqi Niu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Le Xue
- Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, 200040, China
| | - Haotian Wang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Daoyan Hu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310014, China
| | - Yi Liao
- Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, 200040, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Xiaofeng Dou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Congcong Yu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Bo Wang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Yuan Sun
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
- Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, 200040, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310014, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, 310014, China.
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
22
|
Quinn JF, Gray NE. Fluid Biomarkers in Dementia Diagnosis. Continuum (Minneap Minn) 2024; 30:1790-1800. [PMID: 39620844 DOI: 10.1212/con.0000000000001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This article familiarizes neurologists with the currently available CSF and plasma biomarkers for the diagnosis of dementia and diagnosis-dependent treatment decisions. LATEST DEVELOPMENTS For Alzheimer disease, the recent US Food and Drug Administration (FDA) approval of monoclonal antibody therapy has increased the urgency of confirming the pathologic diagnosis with biomarkers before initiating therapy. The new availability of disease-modifying therapies also highlights the need for biomarkers to monitor efficacy over time. Both of these needs have been partially addressed by the emergence of improved blood-based biomarkers for Alzheimer disease. Regarding other forms of dementia, the latest development is a CSF assay for aggregated α-synuclein, which permits the biomarker confirmation of synuclein pathology in Lewy body dementia. ESSENTIAL POINTS CSF biomarkers for the diagnosis of Alzheimer disease, Lewy body dementia, and Creutzfeldt-Jakob disease are well established. Blood-based biomarkers for dementia diagnosis are emerging and rapidly evolving. Sensitivity and specificity for diagnosis continue to improve, and they are being incorporated into diagnostic decisions. Fluid biomarkers for monitoring the efficacy of therapy are not yet established. Because serial CSF examinations are impractical, the validation of blood-based biomarkers of disease activity will be critical for addressing this unmet need.
Collapse
|
23
|
Lee SS, Civitelli L, Parkkinen L. Brain-derived and in vitro-seeded alpha-synuclein fibrils exhibit distinct biophysical profiles. eLife 2024; 13:RP92775. [PMID: 39584804 PMCID: PMC11588339 DOI: 10.7554/elife.92775] [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] [Indexed: 11/26/2024] Open
Abstract
The alpha-synuclein (αSyn) seeding amplification assay (SAA) that allows the generation of disease-specific in vitro seeded fibrils (SAA fibrils) is used as a research tool to study the connection between the structure of αSyn fibrils, cellular seeding/spreading, and the clinicopathological manifestations of different synucleinopathies. However, structural differences between human brain-derived and SAA αSyn fibrils have been recently highlighted. Here, we characterize the biophysical properties of the human brain-derived αSyn fibrils from the brains of patients with Parkinson's disease with and without dementia (PD, PDD), dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and compare them to the 'model' SAA fibrils. We report that the brain-derived αSyn fibrils show distinct biochemical profiles, which were not replicated in the corresponding SAA fibrils. Furthermore, the brain-derived αSyn fibrils from all synucleinopathies displayed a mixture of 'straight' and 'twisted' microscopic structures. However, the PD, PDD, and DLB SAA fibrils had a 'straight' structure, whereas MSA SAA fibrils showed a 'twisted' structure. Finally, the brain-derived αSyn fibrils from all four synucleinopathies were phosphorylated (S129). Interestingly, phosphorylated αSyn were carried over to the PDD and DLB SAA fibrils. Our findings demonstrate the limitation of the SAA fibrils modeling the brain-derived αSyn fibrils and pay attention to the necessity of deepening the understanding of the SAA fibrillation methodology.
Collapse
Affiliation(s)
- Selene Seoyun Lee
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| | - Livia Civitelli
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
24
|
Böing C, Di Fabrizio M, Burger D, Bol JGJM, Huisman E, Rozemuller AJM, van de Berg WDJ, Stahlberg H, Lewis AJ. Distinct ultrastructural phenotypes of glial and neuronal alpha-synuclein inclusions in multiple system atrophy. Brain 2024; 147:3727-3741. [PMID: 38696728 PMCID: PMC11531854 DOI: 10.1093/brain/awae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 03/17/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024] Open
Abstract
Multiple system atrophy is characterized pathologically by the accumulation of alpha-synuclein (aSyn) into glial cytoplasmic inclusions (GCIs). The mechanism underlying the formation of GCIs is not well understood. In this study, correlative light and electron microscopy was employed to investigate aSyn pathology in the substantia nigra and putamen of post-mortem multiple system atrophy brain donors. Three distinct types of aSyn immuno-positive inclusions were identified in oligodendrocytes, neurons and dark cells presumed to be dark microglia. Oligodendrocytes contained fibrillar GCIs that were consistently enriched with lysosomes and peroxisomes, supporting the involvement of the autophagy pathway in aSyn aggregation in multiple system atrophy. Neuronal cytoplasmic inclusions exhibited ultrastructural heterogeneity resembling both fibrillar and membranous inclusions, linking multiple systems atrophy and Parkinson's disease. The novel aSyn pathology identified in the dark cells, displayed GCI-like fibrils or non-GCI-like ultrastructures suggesting various stages of aSyn accumulation in these cells. The observation of GCI-like fibrils within dark cells suggests these cells may be an important contributor to the origin or spread of pathological aSyn in multiple system atrophy. Our results suggest a complex interplay between multiple cell types that may underlie the formation of aSyn pathology in multiple system atrophy brain and highlight the need for further investigation into cell-specific disease pathologies in multiple system atrophy.
Collapse
Affiliation(s)
- Carolin Böing
- C-CINA, Biozentrum, University of Basel, Basel 4058, Switzerland
| | - Marta Di Fabrizio
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Federale Lausanne, Lausanne, Vaud 1015, Switzerland
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud 1015, Switzerland
| | - Domenic Burger
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Federale Lausanne, Lausanne, Vaud 1015, Switzerland
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud 1015, Switzerland
| | - John G J M Bol
- Department of Anatomy and Neurosciences, section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
| | - Evelien Huisman
- Department of Anatomy and Neurosciences, section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
- Amsterdam Neuroscience program Neurodegeneration, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
- Amsterdam Neuroscience program Neurodegeneration, Amsterdam University Medical Centre, Vrije University Amsterdam, Amsterdam 1081 HZ, The Netherlands
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Federale Lausanne, Lausanne, Vaud 1015, Switzerland
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud 1015, Switzerland
| | - Amanda J Lewis
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Federale Lausanne, Lausanne, Vaud 1015, Switzerland
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud 1015, Switzerland
| |
Collapse
|
25
|
Sokratian A, Zhou Y, Tatli M, Burbidge KJ, Xu E, Viverette E, Donzelli S, Duda AM, Yuan Y, Li H, Strader S, Patel N, Shiell L, Malankhanova T, Chen O, Mazzulli JR, Perera L, Stahlberg H, Borgnia M, Bartesaghi A, Lashuel HA, West AB. Mouse α-synuclein fibrils are structurally and functionally distinct from human fibrils associated with Lewy body diseases. SCIENCE ADVANCES 2024; 10:eadq3539. [PMID: 39485845 PMCID: PMC11800946 DOI: 10.1126/sciadv.adq3539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024]
Abstract
The intricate process of α-synuclein aggregation and fibrillization holds pivotal roles in Parkinson's disease (PD) and multiple system atrophy (MSA). While mouse α-synuclein can fibrillize in vitro, whether these fibrils commonly used in research to induce this process or form can reproduce structures in the human brain remains unknown. Here, we report the first atomic structure of mouse α-synuclein fibrils, which was solved in parallel by two independent teams. The structure shows striking similarity to MSA-amplified and PD-associated E46K fibrils. However, mouse α-synuclein fibrils display altered packing arrangements, reduced hydrophobicity, and heightened fragmentation sensitivity and evoke only weak immunological responses. Furthermore, mouse α-synuclein fibrils exhibit exacerbated pathological spread in neurons and humanized α-synuclein mice. These findings provide critical insights into the structural underpinnings of α-synuclein pathogenicity and emphasize a need to reassess the role of mouse α-synuclein fibrils in the development of related diagnostic probes and therapeutic interventions.
Collapse
Affiliation(s)
- Arpine Sokratian
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Ye Zhou
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Meltem Tatli
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, and Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Kevin J. Burbidge
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Enquan Xu
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Elizabeth Viverette
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Sonia Donzelli
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Addison M. Duda
- Department of Chemistry, Duke University, Durham, NC 27708, USA
| | - Yuan Yuan
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Huizhong Li
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Samuel Strader
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Nirali Patel
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Lauren Shiell
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Tuyana Malankhanova
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Olivia Chen
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Joseph R. Mazzulli
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lalith Perera
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, and Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Mario Borgnia
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Alberto Bartesaghi
- Department of Computer Science, Duke University, Durham, NC 27708, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27705, USA
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27708, USA
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Qatar Foundation ND BioSciences, Qatar Foundation Headquarters, PO Box 3400, Al Rayyan, Qatar
| | - Andrew B. West
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
26
|
Gadhave K, Xu E, Wang N, Zhang X, Deyell J, Yang J, Wang A, Cha Y, Kumbhar R, Liu H, Niu L, Chen R, Zhang S, Bakker CC, Jin L, Liang Y, Ying M, Dawson VL, Dawson TM, Rosenthal LS, Mao X. α-Synuclein Strain Dynamics Correlate with Cognitive Shifts in Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619694. [PMID: 39484387 PMCID: PMC11526871 DOI: 10.1101/2024.10.22.619694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
α-Synuclein (α-syn) strains can serve as discriminators between Parkinson's disease (PD) from other α-synucleinopathies. The relationship between α-syn strain dynamics and clinical performance as patients transition from normal cognition (NC) to cognitive impairment (CI) is not known. Here, we show that the biophysical properties and neurotoxicity of α-syn strains change as PD cognitive status transitions from NC to mild cognitive impairment (PD-MCI) and dementia (PD-D). Both cross-sectional and longitudinal analyses reveal distinct α-syn strains in PD patients correlating to their level of cognitive impairment. This study presents evidence that individuals with PD have different α-syn strains that change in accordance with their cognitive status and highlights the potential of α-syn strain dynamics to guide future diagnosis, management, and stratification of PD patients.
Collapse
Affiliation(s)
- Kundlik Gadhave
- 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
| | - Ning 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
| | - Xiaodi Zhang
- 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
| | - Jacob Deyell
- 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
| | - Jun Yang
- 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
| | - Anthony 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
| | - Youngjae Cha
- 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
| | - Ramhari Kumbhar
- 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
| | - Haiqing Liu
- 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
| | - Lili Niu
- 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
| | - Rong Chen
- 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
| | - Shu Zhang
- 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
| | - Catherine C. Bakker
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lingtao Jin
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mingyao Ying
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Hugo W. Moser Research Institute at Kennedy Krieger, 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
| | - 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
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Liana S. Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, 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
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
27
|
Luo H, Yu X, Li P, Hu J, Li W, Li X, Chen M, Yu S. Different neurotoxicity and seeding activity between α-synuclein oligomers formed in plasma of patients with Parkinson's disease and multiple system atrophy. Neuroscience 2024; 557:1-11. [PMID: 39127345 DOI: 10.1016/j.neuroscience.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Previous studies have shown that α-synuclein (α-Syn) aggregates derived from the brains of patients with Parkinson's disease (PD) and multiple system atrophy (MSA) exhibit different phosphorylation, cytotoxicity, and seeding activity. However, the mechanism underlying the differences remains poorly understood. Here, recombinant human α-Syn was incubated in the plasma of patients with PD and MSA, and the oligomers formed in the plasma (PD-O-α-Syn and MSA-O-α-Syn) were purified and analyzed for their phosphorylation, cytotoxicity and seeding activity. In vitro assays revealed that both PD-O-α-Syn and MSA-O-α-Syn were phosphorylated at serine 129. However, the phosphorylation degree of MSA-O-α-Syn was significantly higher than that of PD-O-α-Syn. In addition, MSA-O-α-Syn exhibited stronger cytotoxicity and seeding activity compared with PD-O-α-Syn. In vivo experiments showed that mice receiving intrastriatal inoculation of MSA-O-α-Syn developed more severe motor dysfunction and dopaminergic degeneration than mice receiving intrastriatal inoculation of PD-O-α-Syn. Compared with the mice inoculated with PD-O-α-Syn, the mice inoculated with MSA-O-α-Syn accumulated more phosphorylated and oligomerized α-Syn in the striatum and brain regions (substantia nigra, hippocampus and prefrontal cortex) away from the inoculated site. The results obtained suggest that α-Syn oligomers formed in PD and MSA plasma are different in phosphorylation, cytotoxicity, and seeding activity.
Collapse
Affiliation(s)
- Hanjiang Luo
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Province, China
| | - Xiaohan Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Pengjie Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Junya Hu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Wei Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xin Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Min Chen
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Province, China.
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; National Clinical Research Center for Geriatric Disorders, Beijing 100053, China.
| |
Collapse
|
28
|
Lau D, Tang Y, Kenche V, Copie T, Kempe D, Jary E, Graves NJ, Biro M, Masters CL, Dzamko N, Gambin Y, Sierecki E. Single-Molecule Fingerprinting Reveals Different Growth Mechanisms in Seed Amplification Assays for Different Polymorphs of α-Synuclein Fibrils. ACS Chem Neurosci 2024; 15:3270-3285. [PMID: 39197832 PMCID: PMC11413846 DOI: 10.1021/acschemneuro.4c00185] [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: 03/28/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 09/01/2024] Open
Abstract
α-Synuclein (αSyn) aggregates, detected in the biofluids of patients with Parkinson's disease (PD), have the ability to catalyze their own aggregation, leading to an increase in the number and size of aggregates. This self-templated amplification is used by newly developed assays to diagnose Parkinson's disease and turns the presence of αSyn aggregates into a biomarker of the disease. It has become evident that αSyn can form fibrils with slightly different structures, called "strains" or polymorphs, but little is known about their differential reactivity in diagnostic assays. Here, we compared the properties of two well-described αSyn polymorphs. Using single-molecule techniques, we observed that one of the polymorphs had an increased tendency to undergo secondary nucleation and we showed that this could explain the differences in reactivity observed in in vitro seed amplification assay and cellular assays. Simulations and high-resolution microscopy suggest that a 100-fold difference in the apparent rate of growth can be generated by a surprisingly low number of secondary nucleation "points" (1 every 2000 monomers added by elongation). When both strains are present in the same seeded reaction, secondary nucleation displaces proportions dramatically and causes a single strain to dominate the reaction as the major end product.
Collapse
Affiliation(s)
- Derrick Lau
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Yuan Tang
- Brain
and Mind Centre and Faculty of Medicine and Health, School of Medical
Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Vijaya Kenche
- Florey
Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Thomas Copie
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Daryan Kempe
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Eve Jary
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Noah J. Graves
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Maté Biro
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Colin L. Masters
- Florey
Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicolas Dzamko
- Brain
and Mind Centre and Faculty of Medicine and Health, School of Medical
Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Yann Gambin
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| | - Emma Sierecki
- EMBL
Australia Node for Single Molecule Science and School of Biomedical
Sciences, Faculty of Medicine, The University
of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
29
|
Kannarkat GT, Zack R, Skrinak RT, Morley JF, Davila-Rivera R, Arezoumandan S, Dorfmann K, Luk K, Wolk DA, Weintraub D, Tropea TF, Lee EB, Xie SX, Chandrasekaran G, Lee VMY, Irwin D, Akhtar RS, Chen-Plotkin AS. α-Synuclein Conformations in Plasma Distinguish Parkinson's Disease from Dementia with Lewy Bodies. RESEARCH SQUARE 2024:rs.3.rs-5033901. [PMID: 39372921 PMCID: PMC11451739 DOI: 10.21203/rs.3.rs-5033901/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Aggregation of misfolded α-synuclein (aSyn) within the brain is the pathologic hallmark of Lewy body diseases (LBD), including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Evidence exists for aSyn "strains" - conformations with distinct biological properties. However, biomarkers for PD vs. DLB, including potential aSyn strain differences, are lacking. Here, we used two monoclonal antibodies selective for different in vitro aSyn species - termed Strain A and B - to evaluate human brain tissue, cerebrospinal fluid (CSF), and plasma. Surprisingly, levels of Strain A and B aSyn species differed in plasma from individuals with PD vs. DLB in two independent cohorts. Lower plasma aSyn Strain A species also predicted subsequent PD cognitive decline. Strain A and Strain B aSyn species were undetectable in CSF, but plasma aSyn species could template aSyn fibrillization, particularly in PD. Our findings suggest that aSyn strains may impact LBD clinical presentation and originate outside the brain.
Collapse
Affiliation(s)
- George T. Kannarkat
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Rebecca Zack
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - R. Tyler Skrinak
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - James F. Morley
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
- Parkinson’s Disease Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center; Philadelphia, PA, USA, 19104
| | - Roseanne Davila-Rivera
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Sanaz Arezoumandan
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Katherine Dorfmann
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - David A. Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Daniel Weintraub
- Parkinson’s Disease Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center; Philadelphia, PA, USA, 19104
- Department of Psychiatry and Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Thomas F. Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Edward B. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Sharon X. Xie
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Ganesh Chandrasekaran
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Virginia M.-Y. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - David Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| | - Rizwan S. Akhtar
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, 60611
| | - Alice S. Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA, 19104
| |
Collapse
|
30
|
Lam I, Ndayisaba A, Lewis AJ, Fu Y, Sagredo GT, Kuzkina A, Zaccagnini L, Celikag M, Sandoe J, Sanz RL, Vahdatshoar A, Martin TD, Morshed N, Ichihashi T, Tripathi A, Ramalingam N, Oettgen-Suazo C, Bartels T, Boussouf M, Schäbinger M, Hallacli E, Jiang X, Verma A, Tea C, Wang Z, Hakozaki H, Yu X, Hyles K, Park C, Wang X, Theunissen TW, Wang H, Jaenisch R, Lindquist S, Stevens B, Stefanova N, Wenning G, van de Berg WDJ, Luk KC, Sanchez-Pernaute R, Gómez-Esteban JC, Felsky D, Kiyota Y, Sahni N, Yi SS, Chung CY, Stahlberg H, Ferrer I, Schöneberg J, Elledge SJ, Dettmer U, Halliday GM, Bartels T, Khurana V. Rapid iPSC inclusionopathy models shed light on formation, consequence, and molecular subtype of α-synuclein inclusions. Neuron 2024; 112:2886-2909.e16. [PMID: 39079530 PMCID: PMC11377155 DOI: 10.1016/j.neuron.2024.06.002] [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: 11/29/2022] [Revised: 10/26/2023] [Accepted: 06/03/2024] [Indexed: 09/07/2024]
Abstract
The heterogeneity of protein-rich inclusions and its significance in neurodegeneration is poorly understood. Standard patient-derived iPSC models develop inclusions neither reproducibly nor in a reasonable time frame. Here, we developed screenable iPSC "inclusionopathy" models utilizing piggyBac or targeted transgenes to rapidly induce CNS cells that express aggregation-prone proteins at brain-like levels. Inclusions and their effects on cell survival were trackable at single-inclusion resolution. Exemplar cortical neuron α-synuclein inclusionopathy models were engineered through transgenic expression of α-synuclein mutant forms or exogenous seeding with fibrils. We identified multiple inclusion classes, including neuroprotective p62-positive inclusions versus dynamic and neurotoxic lipid-rich inclusions, both identified in patient brains. Fusion events between these inclusion subtypes altered neuronal survival. Proteome-scale α-synuclein genetic- and physical-interaction screens pinpointed candidate RNA-processing and actin-cytoskeleton-modulator proteins like RhoA whose sequestration into inclusions could enhance toxicity. These tractable CNS models should prove useful in functional genomic analysis and drug development for proteinopathies.
Collapse
Affiliation(s)
- Isabel Lam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Alain Ndayisaba
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Amanda J Lewis
- École Polytechnique Fédérale de Lausanne and University of Lausanne, Lausanne, Switzerland
| | - YuHong Fu
- The University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Science, Sydney, NSW, Australia
| | - Giselle T Sagredo
- The University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Science, Sydney, NSW, Australia
| | - Anastasia Kuzkina
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Meral Celikag
- Dementia Research Institute, University College London, London, UK
| | - Jackson Sandoe
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Ricardo L Sanz
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Aazam Vahdatshoar
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Timothy D Martin
- Harvard Medical School, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Nader Morshed
- Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Boston Children's Hospital, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Arati Tripathi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Charlotte Oettgen-Suazo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Theresa Bartels
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Manel Boussouf
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Max Schäbinger
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Erinc Hallacli
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Xin Jiang
- Yumanity Therapeutics, Cambridge, MA, USA
| | - Amrita Verma
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Challana Tea
- University of California, San Diego, San Diego, CA, USA
| | - Zichen Wang
- University of California, San Diego, San Diego, CA, USA
| | | | - Xiao Yu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Kelly Hyles
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Chansaem Park
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Xinyuan Wang
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Haoyi Wang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Beth Stevens
- Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Boston Children's Hospital, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Kelvin C Luk
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rosario Sanchez-Pernaute
- BioBizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | | | - Daniel Felsky
- Centre for Addiction and Mental Health, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada
| | | | - Nidhi Sahni
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA
| | - S Stephen Yi
- The University of Texas at Austin, Austin, TX, USA
| | | | - Henning Stahlberg
- École Polytechnique Fédérale de Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Isidro Ferrer
- The University of Barcelona, Institut d'Investigacio Biomedica de Bellvitge IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | | | - Stephen J Elledge
- Harvard Medical School, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Glenda M Halliday
- The University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Science, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Tim Bartels
- Dementia Research Institute, University College London, London, UK
| | - Vikram Khurana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
31
|
Frey L, Ghosh D, Qureshi BM, Rhyner D, Guerrero-Ferreira R, Pokharna A, Kwiatkowski W, Serdiuk T, Picotti P, Riek R, Greenwald J. On the pH-dependence of α-synuclein amyloid polymorphism and the role of secondary nucleation in seed-based amyloid propagation. eLife 2024; 12:RP93562. [PMID: 39196271 PMCID: PMC11357353 DOI: 10.7554/elife.93562] [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] [Indexed: 08/29/2024] Open
Abstract
The aggregation of the protein α-synuclein is closely associated with several neurodegenerative disorders and as such the structures of the amyloid fibril aggregates have high scientific and medical significance. However, there are dozens of unique atomic-resolution structures of these aggregates, and such a highly polymorphic nature of the α-synuclein fibrils hampers efforts in disease-relevant in vitro studies on α-synuclein amyloid aggregation. In order to better understand the factors that affect polymorph selection, we studied the structures of α-synuclein fibrils in vitro as a function of pH and buffer using cryo-EM helical reconstruction. We find that in the physiological range of pH 5.8-7.4, a pH-dependent selection between Type 1, 2, and 3 polymorphs occurs. Our results indicate that even in the presence of seeds, the polymorph selection during aggregation is highly dependent on the buffer conditions, attributed to the non-polymorph-specific nature of secondary nucleation. We also uncovered two new polymorphs that occur at pH 7.0 in phosphate-buffered saline. The first is a monofilament Type 1 fibril that highly resembles the structure of the juvenile-onset synucleinopathy polymorph found in patient-derived material. The second is a new Type 5 polymorph that resembles a polymorph that has been recently reported in a study that used diseased tissues to seed aggregation. Taken together, our results highlight the shallow amyloid energy hypersurface that can be altered by subtle changes in the environment, including the pH which is shown to play a major role in polymorph selection and in many cases appears to be the determining factor in seeded aggregation. The results also suggest the possibility of producing disease-relevant structure in vitro.
Collapse
Affiliation(s)
- Lukas Frey
- Institute of Molecular Physical ScienceZürichSwitzerland
| | - Dhiman Ghosh
- Institute of Molecular Physical ScienceZürichSwitzerland
| | - Bilal M Qureshi
- Scientific Center for Optical and Electron MicroscopyZürichSwitzerland
| | - David Rhyner
- Institute of Molecular Physical ScienceZürichSwitzerland
| | | | | | | | - Tetiana Serdiuk
- Institute of Molecular Systems Biology, ETH ZürichZurichSwitzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, ETH ZürichZurichSwitzerland
| | - Roland Riek
- Institute of Molecular Physical ScienceZürichSwitzerland
| | | |
Collapse
|
32
|
Bouvier-Müller A, Fourmy D, Fenyi A, Bousset L, Melki R, Ducongé F. Aptamer binding footprints discriminate α-synuclein fibrillar polymorphs from different synucleinopathies. Nucleic Acids Res 2024; 52:8072-8085. [PMID: 38917326 PMCID: PMC11317169 DOI: 10.1093/nar/gkae544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/02/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Synucleinopathies, including dementia with Lewy bodies (DLB), Parkinson's disease (PD), and multiple system atrophy (MSA), are characterized by the presence of α-synuclein (α-syn) aggregates in the central nervous system. Recent evidence suggests that the heterogeneity of synucleinopathies may be partly explained by the fact that patients may have different α-syn fibrillar polymorphs with structural differences. In this study, we identify nuclease resistant 2'fluoro-pyrimidine RNA aptamers that can differentially bind to structurally distinct α-syn fibrillar polymorphs. Moreover, we introduce a method, AptaFOOT-Seq, designed to rapidly assess the affinity of a mixture of these aptamers for different α-SYN fibrillar polymorphs using next-generation sequencing. Our findings reveal that the binding behavior of aptamers can be very different when they are tested separately or in the presence of other aptamers. In this case, competition and cooperation can occur, providing a higher level of information, which can be exploited to obtain specific 'footprints' for different α-Syn fibrillar polymorphs. Notably, these footprints can distinguish polymorphs obtained from patients with PD, DLB or MSA. This result suggests that aptaFOOT-Seq could be used for the detection of misfolded or abnormal protein conformations to improve the diagnosis of synucleinopathies.
Collapse
Affiliation(s)
- Alix Bouvier-Müller
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| | - Deborah Fourmy
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| | - Alexis Fenyi
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| | - Luc Bousset
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| | - Ronald Melki
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| | - Frédéric Ducongé
- CEA, DRF, Institut of biology JACOB, Molecular Imaging Research Center (MIRCen), Fontenay aux roses 92335, France
- CNRS UMR 9199, Laboratoire des Maladies Neurodégénératives, Fontenay aux roses 92335, France
- Université Paris-Saclay, Fontenay aux roses 92335, France
| |
Collapse
|
33
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [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: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
34
|
Staerz SD, Anamoah C, Tepe JJ. 20S proteasome enhancers prevent cytotoxic tubulin polymerization-promoting protein induced α-synuclein aggregation. iScience 2024; 27:110166. [PMID: 38974969 PMCID: PMC11225362 DOI: 10.1016/j.isci.2024.110166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Synucleinopathies are a class of neurodegenerative diseases defined by the presence of α-synuclein inclusions. The location and composition of these α-synuclein inclusions directly correlate to the disease pattern. The inclusions in Multiple System Atrophy are located predominantly in oligodendrocytes and are rich in a second protein, p25α. P25α plays a key role in neuronal myelination by oligodendrocytes. In healthy oligodendrocytes, there is little to no α-synuclein present. If aberrant α-synuclein is present, p25α leaves the myelin sheaths and quickly co-aggregates with α-synuclein, resulting in the disruption of the cellular process and ultimately cell death. Herein, we report that p25α is susceptible for 20S proteasome-mediated degradation and that p25α induces α-synuclein aggregation, resulting in proteasome impairment and cell death. In addition, we identified small molecules 20S proteasome enhancers that prevent p25α induced α-synuclein fibrilization, restore proteasome impairment, and enhance cell viability.
Collapse
Affiliation(s)
- Sophia D. Staerz
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Charles Anamoah
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Jetze J. Tepe
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|
35
|
Todd TW, Islam NN, Cook CN, Caulfield TR, Petrucelli L. Cryo-EM structures of pathogenic fibrils and their impact on neurodegenerative disease research. Neuron 2024; 112:2269-2288. [PMID: 38834068 PMCID: PMC11257806 DOI: 10.1016/j.neuron.2024.05.012] [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: 08/22/2023] [Revised: 03/13/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024]
Abstract
Neurodegenerative diseases are commonly associated with the formation of aberrant protein aggregates within the brain, and ultrastructural analyses have revealed that the proteins within these inclusions often assemble into amyloid filaments. Cryoelectron microscopy (cryo-EM) has emerged as an effective method for determining the near-atomic structure of these disease-associated filamentous proteins, and the resulting structures have revolutionized the way we think about aberrant protein aggregation and propagation during disease progression. These structures have also revealed that individual fibril conformations may dictate different disease conditions, and this newfound knowledge has improved disease modeling in the lab and advanced the ongoing pursuit of clinical tools capable of distinguishing and targeting different pathogenic entities within living patients. In this review, we summarize some of the recently developed cryo-EM structures of ex vivo α-synuclein, tau, β-amyloid (Aβ), TAR DNA-binding protein 43 (TDP-43), and transmembrane protein 106B (TMEM106B) fibrils and discuss how these structures are being leveraged toward mechanistic research and therapeutic development.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Naeyma N Islam
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Casey N Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
Woerman AL, Bartz JC. Effect of host and strain factors on α-synuclein prion pathogenesis. Trends Neurosci 2024; 47:538-550. [PMID: 38806297 PMCID: PMC11236502 DOI: 10.1016/j.tins.2024.05.004] [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: 02/27/2024] [Revised: 04/19/2024] [Accepted: 05/04/2024] [Indexed: 05/30/2024]
Abstract
Prion diseases are a group of neurodegenerative disorders caused by misfolding of proteins into pathogenic conformations that self-template to spread disease. Although this mechanism is largely associated with the prion protein (PrP) in classical prion diseases, a growing literature indicates that other proteins, including α-synuclein, rely on a similar disease mechanism. Notably, α-synuclein misfolds into distinct conformations, or strains, that cause discrete clinical disorders including multiple system atrophy (MSA) and Parkinson's disease (PD). Because the recognized similarities between PrP and α-synuclein are increasing, this review article draws from research on PrP to identify the host and strain factors that impact disease pathogenesis, predominantly in rodent models, and focuses on key considerations for future research on α-synuclein prions.
Collapse
Affiliation(s)
- Amanda L Woerman
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, USA.
| | - Jason C Bartz
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, USA; Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, USA.
| |
Collapse
|
37
|
Soto C. α-Synuclein seed amplification technology for Parkinson's disease and related synucleinopathies. Trends Biotechnol 2024; 42:829-841. [PMID: 38395703 PMCID: PMC11223967 DOI: 10.1016/j.tibtech.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
Synucleinopathies are a group of neurodegenerative diseases (NDs) associated with cerebral accumulation of α-synuclein (αSyn) misfolded aggregates. At this time, there is no effective treatment to stop or slow down disease progression, which in part is due to the lack of an early and objective biochemical diagnosis. In the past 5 years, the seed amplification technology has emerged for highly sensitive identification of these diseases, even at the preclinical stage of the illness. Much research has been done in multiple laboratories to validate the efficacy and reproducibility of this assay. This article provides a comprehensive review of this technology, including its conceptual basis and its multiple applications for disease diagnosis, as well for understanding of the disease biology and therapeutic development.
Collapse
Affiliation(s)
- Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, TX77030, USA.
| |
Collapse
|
38
|
Yang Y, Zhang Z. α-Synuclein pathology from the body to the brain: so many seeds so close to the central soil. Neural Regen Res 2024; 19:1463-1472. [PMID: 38051888 PMCID: PMC10883481 DOI: 10.4103/1673-5374.387967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/24/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT α-Synuclein is a protein that mainly exists in the presynaptic terminals. Abnormal folding and accumulation of α-synuclein are found in several neurodegenerative diseases, including Parkinson's disease. Aggregated and highly phosphorylated α-synuclein constitutes the main component of Lewy bodies in the brain, the pathological hallmark of Parkinson's disease. For decades, much attention has been focused on the accumulation of α-synuclein in the brain parenchyma rather than considering Parkinson's disease as a systemic disease. Recent evidence demonstrates that, at least in some patients, the initial α-synuclein pathology originates in the peripheral organs and spreads to the brain. Injection of α-synuclein preformed fibrils into the gastrointestinal tract triggers the gut-to-brain propagation of α-synuclein pathology. However, whether α-synuclein pathology can occur spontaneously in peripheral organs independent of exogenous α-synuclein preformed fibrils or pathological α-synuclein leakage from the central nervous system remains under investigation. In this review, we aimed to summarize the role of peripheral α-synuclein pathology in the pathogenesis of Parkinson's disease. We also discuss the pathways by which α-synuclein pathology spreads from the body to the brain.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Lloyd GM, Quintin S, Sorrentino ZA, Gorion KMM, Bell BM, Long B, Paterno G, Giasson BI. A multiverse of α-synuclein: investigation of prion strain properties with carboxyl-terminal truncation specific antibodies in animal models. Acta Neuropathol Commun 2024; 12:91. [PMID: 38858742 PMCID: PMC11163735 DOI: 10.1186/s40478-024-01805-z] [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: 03/29/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders characterized by the presence of misfolded α-Synuclein (αSyn) in the brain. These conditions manifest with diverse clinical and pathophysiological characteristics. This disease diversity is hypothesized to be driven by αSyn strains with differing biophysical properties, potentially influencing prion-type propagation and consequentially the progression of illness. Previously, we investigated this hypothesis by injecting brain lysate (seeds) from deceased individuals with various synucleinopathies or human recombinant αSyn preformed fibrils (PFFs) into transgenic mice overexpressing either wild type or A53T human αSyn. In the studies herein, we expanded on these experiments, utilizing a panel of antibodies specific for the major carboxyl-terminally truncated forms of αSyn (αSynΔC). These modified forms of αSyn are found enriched in human disease brains to inform on potential strain-specific proteolytic patterns. With monoclonal antibodies specific for human αSyn cleaved at residues 103, 114, 122, 125, and 129, we demonstrate that multiple system atrophy (MSA) seeds and PFFs induce differing neuroanatomical spread of αSyn pathology associated with host specific profiles. Overall, αSyn cleaved at residue 103 was most widely present in the induced pathological inclusions. Furthermore, αSynΔC-positive inclusions were present in astrocytes, but more frequently in activated microglia, with patterns dependent on host and inoculum. These findings support the hypothesis that synucleinopathy heterogeneity might stem from αSyn strains with unique biochemical properties that include proteolytic processing, which could result in dominant strain properties.
Collapse
Affiliation(s)
- Grace M Lloyd
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Stephan Quintin
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary A Sorrentino
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Kimberly-Marie M Gorion
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brach M Bell
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Brooke Long
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
| | - Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
40
|
Ishimoto T, Oono M, Kaji S, Ayaki T, Nishida K, Funakawa I, Maki T, Matsuzawa SI, Takahashi R, Yamakado H. A novel mouse model for investigating α-synuclein aggregates in oligodendrocytes: implications for the glial cytoplasmic inclusions in multiple system atrophy. Mol Brain 2024; 17:28. [PMID: 38790036 PMCID: PMC11127389 DOI: 10.1186/s13041-024-01104-7] [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: 12/18/2023] [Accepted: 05/20/2024] [Indexed: 05/26/2024] Open
Abstract
The aggregated alpha-synuclein (αsyn) in oligodendrocytes (OLGs) is one of the pathological hallmarks in multiple system atrophy (MSA). We have previously reported that αsyn accumulates not only in neurons but also in OLGs long after the administration of αsyn preformed fibrils (PFFs) in mice. However, detailed spatial and temporal analysis of oligodendroglial αsyn aggregates was technically difficult due to the background neuronal αsyn aggregates. The aim of this study is to create a novel mouse that easily enables sensitive and specific detection of αsyn aggregates in OLGs and the comparable analysis of the cellular tropism of αsyn aggregates in MSA brains. To this end, we generated transgenic (Tg) mice expressing human αsyn-green fluorescent protein (GFP) fusion proteins in OLGs under the control of the 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter (CNP-SNCAGFP Tg mice). Injection of αsyn PFFs in these mice induced distinct GFP-positive aggregates in the processes of OLGs as early as one month post-inoculation (mpi), and their number and size increased in a centripetal manner. Moreover, MSA-brain homogenates (BH) induced significantly more oligodendroglial αsyn aggregates than neuronal αsyn aggregates compared to DLB-BH in CNP-SNCAGFP Tg mice, suggestive of their potential tropism of αsyn seeds for OLGs. In conclusion, CNP-SNCAGFP Tg mice are useful for studying the development and tropism of αsyn aggregates in OLGs and could contribute to the development of therapeutics targeting αsyn aggregates in OLGs.
Collapse
Affiliation(s)
- Tomoyuki Ishimoto
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Miki Oono
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Seiji Kaji
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Katsuya Nishida
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital, 1314 Ohara, Sanda, 669-1592, Japan
| | - Itaru Funakawa
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital, 1314 Ohara, Sanda, 669-1592, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Shu-Ichi Matsuzawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Hodaka Yamakado
- Department of Neurology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
41
|
Walton S, Fenyi A, Tittle T, Sidransky E, Pal G, Choi S, Melki R, Killinger BA, Kordower JH. Neither alpha-synuclein fibril strain nor host murine genotype influences seeding efficacy. NPJ Parkinsons Dis 2024; 10:105. [PMID: 38773124 PMCID: PMC11109094 DOI: 10.1038/s41531-024-00679-1] [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: 08/24/2023] [Accepted: 03/07/2024] [Indexed: 05/23/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive motor symptoms and alpha-synuclein (αsyn) aggregation in the nervous system. For unclear reasons, PD patients with certain GBA1 mutations (GBA-PD) have a more aggressive clinical progression. Two testable hypotheses that can potentially account for this phenomenon are that GBA1 mutations promote αsyn spread or drive the generation of highly pathogenic αsyn polymorphs (i.e., strains). We tested these hypotheses by treating homozygous GBA1 D409V knockin (KI) mice with human α-syn-preformed fibrils (PFFs) and treating wild-type mice (WT) with several αsyn-PFF polymorphs amplified from brain autopsy samples collected from patients with idiopathic PD and GBA-PD patients with either homozygous or heterozygous GBA1 mutations. Robust phosphorylated-αsyn (PSER129) positive pathology was observed at the injection site (i.e., the olfactory bulb granule cell layer) and throughout the brain six months following PFF injection. The PFF seeding efficiency and degree of spread were similar regardless of the mouse genotype or PFF polymorphs. We found that PFFs amplified from the human brain, regardless of patient genotype, were generally more effective seeders than wholly synthetic PFFs (i.e., non-amplified); however, PFF concentration differed between these two studies, which might also account for the observed differences. To investigate whether the molecular composition of pathology differed between different seeding conditions, we performed Biotinylation by Antibody Recognition on PSER129 (BAR-PSER129). We found that for BAR-PSER129, the endogenous PSER129 pool dominated identified interactions, and thus, very few potential interactions were explicitly identified for seeded pathology. However, we found Dynactin Subunit 2 (Dctn2) interaction was shared across all PFF conditions, and NCK Associated Protein 1 (Nckap1) and Adaptor Related Protein Complex 3 Subunit Beta 2 (Ap3b2) were unique to PFFs amplified from GBA-PD brains of heterozygous mutation carriers. In conclusion, both the genotype and αsyn strain had little effect on overall seeding efficacy and global PSER129-interactions.
Collapse
Affiliation(s)
- Sara Walton
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Alexis Fenyi
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Tyler Tittle
- Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Gian Pal
- Department of Neurology, Division of Movement Disorders, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Solji Choi
- Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | | | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
42
|
Ullah I, Zhao L, Uddin S, Zhou Y, Wang X, Li H. Nicotine-mediated therapy for Parkinson's disease in transgenic Caenorhabditis elegans model. Front Aging Neurosci 2024; 16:1358141. [PMID: 38813528 PMCID: PMC11135287 DOI: 10.3389/fnagi.2024.1358141] [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: 12/19/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Parkinson's disease resultant in the degeneration of Dopaminergic neurons and accumulation of α-synuclein in the substantia nigra pars compacta. The synthetic therapeutics for Parkinson's disease have moderate symptomatic benefits but cannot prevent or delay disease progression. In this study, nicotine was employed by using transgenic Caenorhabditis elegans Parkinson's disease models to minimize the Parkinson's disease symptoms. The results showed that the nicotine at 100, 150, and 200 μM doses reduced degeneration of Dopaminergic neurons caused by 6-hydroxydopamine (14, 33, and 40%), lowered the aggregative toxicity of α-synuclein by 53, 56, and 78%, respectively. The reduction in food-sensing behavioral disabilities of BZ555 was observed to be 18, 49, and 86%, respectively, with nicotine concentrations of 100 μM, 150 μM, and 200 μM. Additionally, nicotine was found to enhance Daf-16 nuclear translocation by 14, 31, and 49%, and dose-dependently increased SOD-3 expression by 10, 19, and 23%. In summary, the nicotine might a promising therapy option for Parkinson's disease.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shahab Uddin
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yangtao Zhou
- Department of Neurology, Clinical Center for Parkinson's Disease, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
43
|
Chisholm T, Melki R, Hunter CA. Ligand Profiling as a Diagnostic Tool to Differentiate Patient-Derived α-Synuclein Polymorphs. ACS Chem Neurosci 2024; 15:2080-2088. [PMID: 38690599 PMCID: PMC11099917 DOI: 10.1021/acschemneuro.4c00178] [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: 03/24/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
Amyloid fibrils are characteristic of many neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. While different diseases may have fibrils formed of the same protein, the supramolecular morphology of these fibrils is disease-specific. Here, a method is reported to distinguish eight morphologically distinct amyloid fibrils based on differences in ligand binding properties. Eight fibrillar polymorphs of α-synuclein (αSyn) were investigated: five generated de novo using recombinant αSyn and three generated using protein misfolding cyclic amplification (PMCA) of recombinant αSyn seeded with brain homogenates from deceased patients diagnosed with Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB). Fluorescence binding assays were carried out for each fibril using a toolkit of six different ligands. The fibril samples were separated into five categories based on a binary classification of whether they bound specific ligands or not. Quantitative binding measurements then allowed every fibrillar polymorph to be uniquely identified, and the PMCA fibrils derived from PD, MSA, and DLB patients could be unambiguously distinguished. This approach constitutes a novel and operationally simple method to differentiate amyloid fibril morphologies and to identify disease states using PMCA fibrils obtained by seeding with patient samples.
Collapse
Affiliation(s)
- Timothy
S. Chisholm
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Ronald Melki
- Institut
François Jacob (MIRCen), CEA, CNRS, University Paris-Saclay, 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Christopher A. Hunter
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| |
Collapse
|
44
|
Kannarkat GT, Zack R, Skrinak RT, Morley JF, Davila-Rivera R, Arezoumandan S, Dorfmann K, Luk K, Wolk DA, Weintraub D, Tropea TF, Lee EB, Xie SX, Chandrasekaran G, Lee VMY, Irwin D, Akhtar RS, Chen-Plotkin AS. α-Synuclein Conformations in Plasma Distinguish Parkinson's Disease from Dementia with Lewy Bodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593056. [PMID: 38765963 PMCID: PMC11100683 DOI: 10.1101/2024.05.07.593056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Spread and aggregation of misfolded α-synuclein (aSyn) within the brain is the pathologic hallmark of Lewy body diseases (LBD), including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). While evidence exists for multiple aSyn protein conformations, often termed "strains" for their distinct biological properties, it is unclear whether PD and DLB result from aSyn strain differences, and biomarkers that differentiate PD and DLB are lacking. Moreover, while pathological forms of aSyn have been detected outside the brain ( e.g., in skin, gut, blood), the functional significance of these peripheral aSyn species is unclear. Here, we developed assays using monoclonal antibodies selective for two different aSyn species generated in vitro - termed Strain A and Strain B - and used them to evaluate human brain tissue, cerebrospinal fluid (CSF), and plasma, through immunohistochemistry, enzyme-linked immunoassay, and immunoblotting. Surprisingly, we found that plasma aSyn species detected by these antibodies differentiated individuals with PD vs. DLB in a discovery cohort (UPenn, n=235, AUC 0.83) and a multi-site replication cohort (Parkinson's Disease Biomarker Program, or PDBP, n=200, AUC 0.72). aSyn plasma species detected by the Strain A antibody also predicted rate of cognitive decline in PD. We found no evidence for aSyn strains in CSF, and ability to template aSyn fibrillization differed for species isolated from plasma vs. brain, and in PD vs. DLB. Taken together, our findings suggest that aSyn conformational differences may impact clinical presentation and cortical spread of pathological aSyn. Moreover, the enrichment of these aSyn strains in plasma implicates a non-central nervous system source.
Collapse
|
45
|
Schwab K, Frahm S, Magbagbeolu M, Horsley D, Goatman EA, Melis V, Theuring F, Ishaq A, Storey JMD, Harrington CR, Wischik CM, Riedel G. LETC inhibits α-Syn aggregation and ameliorates motor deficiencies in the L62 mouse model of synucleinopathy. Eur J Pharmacol 2024; 970:176505. [PMID: 38503400 DOI: 10.1016/j.ejphar.2024.176505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
Alpha-Synuclein (α-Syn) aggregation is a pathological feature of synucleinopathies, neurodegenerative disorders that include Parkinson's disease (PD). Here, we explored the efficacy of N,N,N',N'-tetraethyl-10H-phenothiazine-3,7-diamine dihydrochloride (LETC), a protein aggregation inhibitor, on α-Syn aggregation. In both cellular models and transgenic mice, α-Syn aggregation was achieved by the overexpression of full-length human α-Syn fused with a signal sequence peptide. α-Syn accumulated in transfected DH60.21 neuroblastoma cells and α-Syn aggregation was inhibited by LETC with an EC50 of 0.066 ± 0.047 μM. Full-length human α-Syn overexpressing Line 62 (L62) mice accumulated neuronal α-Syn that was associated with a decreased motor performance in the open field and automated home cage. LETC, administered orally for 6 weeks at 10 mg/kg significantly decreased α-Syn-positive neurons in multiple brain regions and this resulted in a rescue of movement deficits in the open field in these mice. LETC however, did not improve activity deficits of L62 mice in the home cage environment. The results suggest that LETC may provide a potential disease modification therapy in synucleinopathies through the inhibition of α-Syn aggregation.
Collapse
Affiliation(s)
- Karima Schwab
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany.
| | - Silke Frahm
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Mandy Magbagbeolu
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - David Horsley
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Elizabeth A Goatman
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Valeria Melis
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Franz Theuring
- Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Ahtsham Ishaq
- Department of Chemistry, University of Aberdeen, Aberdeen, UK
| | - John M D Storey
- Department of Chemistry, University of Aberdeen, Aberdeen, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
46
|
Balana AT, Mahul-Mellier AL, Nguyen BA, Horvath M, Javed A, Hard ER, Jasiqi Y, Singh P, Afrin S, Pedretti R, Singh V, Lee VMY, Luk KC, Saelices L, Lashuel HA, Pratt MR. O-GlcNAc forces an α-synuclein amyloid strain with notably diminished seeding and pathology. Nat Chem Biol 2024; 20:646-655. [PMID: 38347213 PMCID: PMC11062923 DOI: 10.1038/s41589-024-01551-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024]
Abstract
Amyloid-forming proteins such α-synuclein and tau, which are implicated in Alzheimer's and Parkinson's disease, can form different fibril structures or strains with distinct toxic properties, seeding activities and pathology. Understanding the determinants contributing to the formation of different amyloid features could open new avenues for developing disease-specific diagnostics and therapies. Here we report that O-GlcNAc modification of α-synuclein monomers results in the formation of amyloid fibril with distinct core structure, as revealed by cryogenic electron microscopy, and diminished seeding activity in seeding-based neuronal and rodent models of Parkinson's disease. Although the mechanisms underpinning the seeding neutralization activity of the O-GlcNAc-modified fibrils remain unclear, our in vitro mechanistic studies indicate that heat shock proteins interactions with O-GlcNAc fibril inhibit their seeding activity, suggesting that the O-GlcNAc modification may alter the interactome of the α-synuclein fibrils in ways that lead to reduce seeding activity in vivo. Our results show that posttranslational modifications, such as O-GlcNAc modification, of α-synuclein are key determinants of α-synuclein amyloid strains and pathogenicity.
Collapse
Affiliation(s)
- Aaron T Balana
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Anne-Laure Mahul-Mellier
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Binh A Nguyen
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mian Horvath
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Afraah Javed
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Eldon R Hard
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Yllza Jasiqi
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Preeti Singh
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shumaila Afrin
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rose Pedretti
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Virender Singh
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Virginia M-Y Lee
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelvin C Luk
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorena Saelices
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Matthew R Pratt
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
- Department Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
47
|
Makey DM, Gadkari VV, Kennedy RT, Ruotolo BT. Cyclic Ion Mobility-Mass Spectrometry and Tandem Collision Induced Unfolding for Quantification of Elusive Protein Biomarkers. Anal Chem 2024; 96:6021-6029. [PMID: 38557001 PMCID: PMC11081454 DOI: 10.1021/acs.analchem.4c00477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Sensitive analytical techniques that are capable of detecting and quantifying disease-associated biomolecules are indispensable in our efforts to understand disease mechanisms and guide therapeutic intervention through early detection, accurate diagnosis, and effective monitoring of disease. Parkinson's Disease (PD), for example, is one of the most prominent neurodegenerative disorders in the world, but the diagnosis of PD has primarily been based on the observation of clinical symptoms. The protein α-synuclein (α-syn) has emerged as a promising biomarker candidate for PD, but a lack of analytical methods to measure complex disease-associated variants of α-syn has prevented its widespread use as a biomarker. Antibody-based methods such as immunoassays and mass spectrometry-based approaches have been used to measure a limited number of α-syn forms; however, these methods fail to differentiate variants of α-syn that display subtle differences in only the sequence and structure. In this work, we developed a cyclic ion mobility-mass spectrometry method that combines multiple stages of activation and timed ion selection to quantify α-syn variants using both mass- and structure-based measurements. This method can allow for the quantification of several α-syn variants present at physiological levels in biological fluid. Taken together, this approach can be used to galvanize future efforts aimed at understanding the underlying mechanisms of PD and serves as a starting point for the development of future protein-structure-based diagnostics and therapeutic interventions.
Collapse
Affiliation(s)
- Devin M. Makey
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Varun V. Gadkari
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brandon T. Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
48
|
Vovard B, Bodin A, Gouju J, de Guilhem de Lataillade A, Derkinderen P, Etcharry-Bouyx F, Chauviré V, Guillet-Pichon V, Verny C, Letournel F, Lenaers G, Chevrollier A, Codron P. Stochastic Optical Reconstruction Microscopy Imaging of Multiple System Atrophy Inclusions Suggests Stepwise α-Synuclein Aggregation. Mov Disord 2024; 39:723-728. [PMID: 38357858 DOI: 10.1002/mds.29744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The architecture and composition of glial (GCI) and neuronal (NCI) α-synuclein inclusions observed in multiple system atrophy (MSA) remain to be precisely defined to better understand the disease. METHODS Here, we used stochastic optical reconstruction microscopy (STORM) to characterize the nanoscale organization of glial (GCI) and neuronal (NCI) α-synuclein inclusions in cryopreserved brain sections from MSA patients. RESULTS STORM revealed a dense cross-linked internal structure of α-synuclein in all GCI and NCI. The internal architecture of hyperphosphorylated α-synuclein (p-αSyn) inclusions was similar in glial and neuronal cells, suggesting a common aggregation mechanism. A similar sequence of p-αSyn stepwise intracellular aggregation was defined in oligodendrocytes and neurons, starting from the perinuclear area and growing inside the cells. Consistent with this hypothesis, we found a higher mitochondrial density in GCI and NCI compared to oligodendrocytes and neurons from unaffected donors (P < 0.01), suggesting an active recruitment of the organelles during the aggregation process. CONCLUSIONS These first STORM images of GCI and NCI suggest stepwise α-synuclein aggregation in MSA. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Benoît Vovard
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
- Laboratoire de neurobiologie et neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Alexia Bodin
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
- Laboratoire de neurobiologie et neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Julien Gouju
- Laboratoire de neurobiologie et neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France
- MINT, UMR Inserm 1066, CNRS 6021, Angers, France
| | - Adrien de Guilhem de Lataillade
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- Service de Neurologie, CHU Nantes, Inserm U1235 Nantes, Nantes, France
| | - Pascal Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- Service de Neurologie, CHU Nantes, Inserm U1235 Nantes, Nantes, France
| | - Frédérique Etcharry-Bouyx
- Service de Neurologie, centre mémoire de ressource et de recherche, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Valérie Chauviré
- Service de Neurologie, centre mémoire de ressource et de recherche, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Virginie Guillet-Pichon
- Service de Neurologie, centre mémoire de ressource et de recherche, Centre Hospitalier Universitaire d'Angers, Angers, France
- Service de Neurologie, centre de référence des maladies neurogénétiques, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Christophe Verny
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
- Service de Neurologie, centre de référence des maladies neurogénétiques, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Franck Letournel
- Laboratoire de neurobiologie et neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France
- MINT, UMR Inserm 1066, CNRS 6021, Angers, France
| | - Guy Lenaers
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
- Service de Neurologie, centre de référence des maladies neurogénétiques, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Arnaud Chevrollier
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
| | - Philippe Codron
- Univ Angers, Equipe MitoLab, Unité MitoVasc, Inserm U1083, CNRS 6015, SFR ICAT, Angers, France
- Laboratoire de neurobiologie et neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France
| |
Collapse
|
49
|
Abdul‐Rahman T, Herrera‐Calderón RE, Ahluwalia A, Wireko AA, Ferreira T, Tan JK, Wolfson M, Ghosh S, Horbas V, Garg V, Perveen A, Papadakis M, Ashraf GM, Alexiou A. The potential of phosphorylated α-synuclein as a biomarker for the diagnosis and monitoring of multiple system atrophy. CNS Neurosci Ther 2024; 30:e14678. [PMID: 38572788 PMCID: PMC10993367 DOI: 10.1111/cns.14678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 04/05/2024] Open
Abstract
INTRODUCTION Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disorder characterized by the presence of glial cytoplasmic inclusions (GCIs) containing aggregated α-synuclein (α-Syn). Accurate diagnosis and monitoring of MSA present significant challenges, which can lead to potential misdiagnosis and inappropriate treatment. Biomarkers play a crucial role in improving the accuracy of MSA diagnosis, and phosphorylated α-synuclein (p-syn) has emerged as a promising biomarker for aiding in diagnosis and disease monitoring. METHODS A literature search was conducted on PubMed, Scopus, and Google Scholar using specific keywords and MeSH terms without imposing a time limit. Inclusion criteria comprised various study designs including experimental studies, case-control studies, and cohort studies published only in English, while conference abstracts and unpublished sources were excluded. RESULTS Increased levels of p-syn have been observed in various samples from MSA patients, such as red blood cells, cerebrospinal fluid, oral mucosal cells, skin, and colon biopsies, highlighting their diagnostic potential. The α-Syn RT-QuIC assay has shown sensitivity in diagnosing MSA and tracking its progression. Meta-analyses and multicenter investigations have confirmed the diagnostic value of p-syn in cerebrospinal fluid, demonstrating high specificity and sensitivity in distinguishing MSA from other neurodegenerative diseases. Moreover, combining p-syn with other biomarkers has further improved the diagnostic accuracy of MSA. CONCLUSION The p-syn stands out as a promising biomarker for MSA. It is found in oligodendrocytes and shows a correlation with disease severity and progression. However, further research and validation studies are necessary to establish p-syn as a reliable biomarker for MSA. If proven, p-syn could significantly contribute to early diagnosis, disease monitoring, and assessing treatment response.
Collapse
Affiliation(s)
| | | | | | | | - Tomas Ferreira
- Department of Clinical Neurosciences, School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | | | | | - Shankhaneel Ghosh
- Institute of Medical Sciences and SUM Hospital, Siksha 'O' AnusandhanBhubaneswarIndia
| | | | - Vandana Garg
- Department of Pharmaceutical SciencesMaharshi Dayanand UniversityRohtakHaryanaIndia
| | - Asma Perveen
- Glocal School of Life SciencesGlocal UniversitySaharanpurUttar PradeshIndia
- Princess Dr. Najla Bint Saud Al‐Saud Center for Excellence Research in BiotechnologyKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Ghulam Md Ashraf
- Department of Medical Laboratory SciencesUniversity of Sharjah, College of Health Sciences, and Research Institute for Medical and Health SciencesSharjahUAE
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationNew South WalesAustralia
| |
Collapse
|
50
|
Schwab K, Magbagbeolu M, Theuring F, Harrington CR, Wischik CM, Riedel G. Solubility of α-synuclein species in the L62 mouse model of synucleinopathy. Sci Rep 2024; 14:6239. [PMID: 38486089 PMCID: PMC10940722 DOI: 10.1038/s41598-024-56735-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/11/2024] [Indexed: 03/18/2024] Open
Abstract
The accumulation of α-synuclein (α-Syn) into Lewy bodies is a hallmark of synucleinopathies, a group of neurological disorders that include Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Small oligomers as well as larger fibrils of α-Syn have been suggested to induce cell toxicity leading to a degenerative loss of neurones. A richer understanding of α-Syn aggregation in disease, however, requires the identification of the different α-Syn species and the characterisation of their biochemical properties. We here aimed at a more in-depth characterisation of the α-Syn transgenic mice, Line 62 (L62), and examined the deposition pattern and solubility of human and murine α-Syn in these mice using immunohistochemical and biochemical methods. Application of multiple antibodies confirmed mAb syn204 as the most discriminatory antibody for human α-Syn in L62. Syn204 revealed an intense and widespread immunohistochemical α-Syn labelling in parietal cortex and hippocampus, and to a lower level in basal forebrain and hindbrain regions. The labelled α-Syn represented somatic inclusions as well as processes and synaptic endings. Biochemical analysis revealed a Triton-resistant human α-Syn pool of large oligomers, a second pool of small oligomers that was not resistant to solubilization with urea/Triton. A third SDS-soluble pool of intermediate sized aggregates containing a mixture of both, human and mouse α-Syn was also present. These data suggest that several pools of α-Syn can exist in neurones, most likely in different cellular compartments. Information about these different pools is important for the development of novel disease modifying therapies aimed at α-Syn.
Collapse
Affiliation(s)
- Karima Schwab
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Forester Hill, Aberdeen, AB25 2ZD, UK.
- Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany.
| | - Mandy Magbagbeolu
- Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Franz Theuring
- Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Forester Hill, Aberdeen, AB25 2ZD, UK
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Forester Hill, Aberdeen, AB25 2ZD, UK
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen, AB24 5RP, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Forester Hill, Aberdeen, AB25 2ZD, UK
| |
Collapse
|