1
|
Wang Y, Li Y, Gu Y, Ma W, Guan Y, Guo M, Shao Q, Ji X, Liu J. Decreased levels of phosphorylated synuclein in plasma are correlated with poststroke cognitive impairment. Neural Regen Res 2025; 20:2598-2610. [PMID: 38845216 PMCID: PMC11801306 DOI: 10.4103/nrr.nrr-d-23-01348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/30/2023] [Accepted: 02/29/2024] [Indexed: 11/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00022/figure1/v/2024-11-05T132919Z/r/image-tiff Poststroke cognitive impairment is a major secondary effect of ischemic stroke in many patients; however, few options are available for the early diagnosis and treatment of this condition. The aims of this study were to (1) determine the specific relationship between hypoxic and α-synuclein during the occur of poststroke cognitive impairment and (2) assess whether the serum phosphorylated α-synuclein level can be used as a biomarker for poststroke cognitive impairment. We found that the phosphorylated α-synuclein level was significantly increased and showed pathological aggregation around the cerebral infarct area in a mouse model of ischemic stroke. In addition, neuronal α-synuclein phosphorylation and aggregation were observed in the brain tissue of mice subjected to chronic hypoxia, suggesting that hypoxia is the underlying cause of α-synuclein-mediated pathology in the brains of mice with ischemic stroke. Serum phosphorylated α-synuclein levels in patients with ischemic stroke were significantly lower than those in healthy subjects, and were positively correlated with cognition levels in patients with ischemic stroke. Furthermore, a decrease in serum high-density lipoprotein levels in stroke patients was significantly correlated with a decrease in phosphorylated α-synuclein levels. Although ischemic stroke mice did not show significant cognitive impairment or disrupted lipid metabolism 14 days after injury, some of them exhibited decreased cognitive function and reduced phosphorylated α-synuclein levels. Taken together, our results suggest that serum phosphorylated α-synuclein is a potential biomarker for poststroke cognitive impairment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Clinical Laboratory, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Capital Medical University, Beijing, China
| | - Yuning Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuying Guan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Hong JY, Lee JS, Kim SH, Lee PH. A model of L-DOPA-induced dyskinesia in parkinsonian mice produced by AAV vector-mediated overexpression of α-synuclein. Exp Neurol 2025; 389:115264. [PMID: 40239797 DOI: 10.1016/j.expneurol.2025.115264] [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: 11/13/2024] [Revised: 02/26/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
L-3,4-dihydroxyphenylalanin (L-DOPA) is the most effective drug for treating Parkinson's disease (PD); however, long-term L-DOPA therapy can lead to L-DOPA-induced dyskinesia (LID). While the 6-hydroxydopamine-lesioned rodent model for LID fails to reproduce the pathological hallmarks of PD, a newly introduced rodent model using adeno-associated virus (AAV)-mediated overexpression of α-synuclein results in α-synuclein aggregation and progressive loss of dopaminergic neurons. The present study aimed to provoke LID in parkinsonian mice generated by AAV vector-mediated overexpression of α-synuclein and to explore histologic features associated with LID. A recombinant AAV2/7 vector containing the human α-synuclein transgene was injected into the substantia nigra (SN) of wild-type mice. Eight weeks later, mice received daily injections of 10 mg/kg of L-DOPA for one week, followed by 25 mg/kg of L-DOPA daily for the subsequent week. LID was observed in 3 out of 19 mice at the 10 mg/kg L-DOPA dose and in 14 mice at 25 mg/kg dose. The number of tyrosine hydroxylase (TH)-positive neurons in the AAV vector-injected side of the SN was reduced to an average of 59 % of the intact side, and the optical density of TH-positive fibers in the ipsilateral striatum was reduced to an average of 37 %. Abnormal Involuntary Movement scores were correlated with decrease in both the number of TH-positive neurons in SN and optical density of striatal TH-positive fibers. This study establishes a mouse model for LID using AAV vector-mediated overexpression of α-synuclein, providing a useful tool for investigating the progressive changes and associated pathophysiology during occurrence of LID.
Collapse
Affiliation(s)
- Jin Yong Hong
- Department of Neurology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, South Korea.
| | - Jin Suk Lee
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, South Korea
| | - Seo Hyun Kim
- Department of Neurology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, South Korea
| | - Phil Hyu Lee
- Department of Neurology and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
3
|
Brzozowski CF, Challa H, Gcwensa NZ, Hall D, Nabert D, Chambers N, Gallardo I, Millet M, Volpicelli-Daley L, Moehle MS. Early α-synuclein aggregation decreases corticostriatal glutamate drive and synapse density. Neurobiol Dis 2025; 210:106918. [PMID: 40250719 DOI: 10.1016/j.nbd.2025.106918] [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: 01/29/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
Neuronal inclusions of α-synuclein (α-syn) are pathological hallmarks of Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). α-Syn pathology accumulates in cortical neurons which project to the striatum. To understand how α-syn pathology affects cortico-striatal synapses at early time points before significant dopamine neuron loss, pre-formed α-syn fibrils (PFF) were injected into the striatum to induce endogenous α-syn aggregation in corticostriatal-projecting neurons. Electrophysiological recordings of striatal spiny projection neurons (SPNs) from acute slices found a significant decrease in evoked corticostriatal glutamate release and corticostriatal synaptic release sites in mice with PFF-induced aggregates compared to monomer injected mice. Expansion microscopy, confocal microscopy and Imaris reconstructions were used to identify VGLUT1 positive presynaptic terminals juxtaposed to Homer1 positive postsynaptic densities, termed synaptic loci. Quantitation of synaptic loci density revealed an early loss of corticostriatal synapses. Immunoblots of the striatum showed reductions in expression of pre-synaptic proteins VGLUT1, VAMP2 and Snap25, in mice with α-syn aggregates compared to controls. Paradoxically, a small percentage of remaining VGLUT1+ synaptic loci positive for pS129-α-syn aggregates showed enlarged volumes compared to nearby synapses without α-syn aggregates. Our combined physiology and high-resolution imaging data point to an early loss of corticostriatal synapses in mice harboring α-synuclein inclusions, which may contribute to impaired basal ganglia circuitry in PD and DLB.
Collapse
Affiliation(s)
- Charlotte F Brzozowski
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Harshita Challa
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nolwazi Z Gcwensa
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dominic Hall
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Douglas Nabert
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nicole Chambers
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ignacio Gallardo
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Michael Millet
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Laura Volpicelli-Daley
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mark S Moehle
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
4
|
Edwards S, Corrigan F, Collins-Praino L. Lasting Impact: Exploring the Brain Mechanisms that Link Traumatic Brain Injury to Parkinson's Disease. Mol Neurobiol 2025; 62:7421-7444. [PMID: 39891816 PMCID: PMC12078371 DOI: 10.1007/s12035-025-04706-x] [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: 07/22/2024] [Accepted: 01/14/2025] [Indexed: 02/03/2025]
Abstract
Development of Parkinson's Disease (PD) is linked with a history of traumatic brain injury (TBI), although the mechanisms driving this remain unclear. Of note, many key parallels have been identified between the pathologies of PD and TBI; in particular, PD is characterised by loss of dopaminergic neurons from the substantia nigra (SN), accompanied by broader changes to dopaminergic signalling, disruption of the Locus Coeruleus (LC) and noradrenergic system, and accumulation of aggregated α-synuclein in Lewy Bodies, which spreads in a stereotypical pattern throughout the brain. Widespread disruptions to the dopaminergic and noradrenergic systems, including progressive neuronal loss from the SN and LC, have been observed acutely following injury, some of which have also been identified chronically in TBI patients and preclinical models. Furthermore, changes to α-synuclein expression are also seen both acutely and chronically following injury throughout the brain, although detailed characterisation of these changes and spread of pathology is limited. In this review, we detail the current literature regarding dopaminergic and noradrenergic disruption and α-synuclein pathology following injury, with particular focus on how these changes may predispose individuals to prolonged pathology and progressive neurodegeneration, particularly the development of PD. While it is increasingly clear that TBI is a key risk factor for the development of PD, significant gaps remain in current understanding of neurodegenerative pathology following TBI, particularly chronic manifestations of injury.
Collapse
Affiliation(s)
- Samantha Edwards
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
- Head Injury Lab, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Frances Corrigan
- Head Injury Lab, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Lyndsey Collins-Praino
- Cognition, Ageing and Neurodegenerative Disease Laboratory, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
5
|
Mingo YB, Escobar Galvis ML, Henderson MX. α-Synuclein pathology and mitochondrial dysfunction: Toxic partners in Parkinson's disease. Neurobiol Dis 2025; 209:106889. [PMID: 40157617 DOI: 10.1016/j.nbd.2025.106889] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025] Open
Abstract
Two major neuropathological features of Parkinson's disease (PD) are α-synuclein Lewy pathology and mitochondrial dysfunction. Although both α-synuclein pathology and mitochondrial dysfunction may independently contribute to PD pathogenesis, the interaction between these two factors is not yet fully understood. In this review, we discuss the physiological functions of α-synuclein and mitochondrial homeostasis in neurons as well as the pathological defects that ensue when these functions are disturbed in PD. Recent studies have highlighted that dysfunctional mitochondria can become sequestered within Lewy bodies, and cell biology studies have suggested that α-synuclein can directly impair mitochondrial function. There are also PD cases caused by genetic or environmental perturbation of mitochondrial homeostasis. Together, these studies suggest that mitochondrial dysfunction may be a common pathway to neurodegeneration in PD, triggered by multiple insults. We review the literature surrounding the interaction between α-synuclein and mitochondria and highlight open questions in the field that may be explored to advance our understanding of PD and develop novel, disease-modifying therapies.
Collapse
Affiliation(s)
- Yakum B Mingo
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | | | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| |
Collapse
|
6
|
Izco M, Sola C, Schleef M, Schmeer M, de Toro M, Verona G, Carlos E, Reinares-Sebastian A, Colina S, Marzo-Sola ME, Garcia-Sanmartin J, Fernández-Irigoyen J, Santamaría E, Mugica-Vidal R, Blesa J, Alvarez-Erviti L. Development of human targeted extracellular vesicles loaded with shRNA minicircles to prevent parkinsonian pathology. Transl Neurodegener 2025; 14:26. [PMID: 40420149 DOI: 10.1186/s40035-025-00484-7] [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: 11/15/2024] [Accepted: 04/09/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Neurological disorders are the second leading cause of death and the leading cause of disability in the world. Thus, the development of novel disease-modifying strategies is clearly warranted. We have previously developed a therapeutic approach using mouse targeted rabies virus glycoprotein (RVG) extracellular vesicles (EVs) to deliver minicircles (MCs) expressing shRNA (shRNA-MCs) to induce long-term α-synuclein down-regulation. Although the previous therapy successfully reduced the pathology, the clinical translation was extremely unlikely since they were mouse extracellular vesicles. METHODS To overcome this limitation, we developed a source of human RVG-EVs compatible with a personalized therapy using immature dendritic cells. Human peripheral blood monocytes were differentiated in vitro into immature dendritic cells, which were transfected to express the RVG peptide. RVG-EVs containing shRNA-MCs, loaded by electroporation, were injected intravenously in the α-synuclein performed fibril (PFF) mouse model. Level of α-synuclein, phosphorylated α-synuclein aggregates, dopaminergic neurons and motor function were evaluated 90 days after the treatment. To confirm that EVs derived from patients were suitable as a vehicle, proteomic analysis of EVs derived from control, initial and advanced Parkinson's disease was performed. RESULTS The shRNA-MCs could be successfully loaded into human RVG-EVs and downregulate α-synuclein in SH-SY5Y cells. Intravenous injection of the shRNA-MC-loaded RVG-EVs induced long-term downregulation of α-synuclein mRNA expression and protein level, decreased α-synuclein aggregates, prevented dopaminergic cell death and ameliorated motor impairment in the α-synuclein PFF mouse model. Moreover, we confirmed that the EVs from PD patients are suitable as a personalized therapeutic vehicle. CONCLUSION Our study confirmed the therapeutic potential of shRNA-MCs delivered by human RVG-EVs for long-term treatment of neurodegenerative diseases. These results pave the way for clinical use of this approach.
Collapse
Affiliation(s)
- Maria Izco
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain.
| | - Carlos Sola
- Transfusion Center and Blood Bank of La Rioja, 26006, Logroño, Spain
| | | | | | - María de Toro
- Genomics and Bioinformatics Core Facility, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Guglielmo Verona
- Centre for Amyloidosis, UCL Medical School, Rowland Hill Street, London, NW3 2PF, UK
| | - Estefania Carlos
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Alejandro Reinares-Sebastian
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Sandra Colina
- Servicio de Neurología, Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| | | | - Josune Garcia-Sanmartin
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario, Universitario de Navarra (HUN), 31008, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario, Universitario de Navarra (HUN), 31008, Pamplona, Spain
| | - Rodolfo Mugica-Vidal
- Department of Mechanical Engineering, University of La Rioja, 26004, Logroño, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Lydia Alvarez-Erviti
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain.
| |
Collapse
|
7
|
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
|
8
|
Sheng YC, Huang JN, Wu WL, Wan XR, Wang J, Qin ZH, Wang Y. TIGAR plays neuroprotective roles in MPP +/MPTP-induced Parkinson's disease by alleviating ferroptosis. Eur J Pharmacol 2025; 995:177430. [PMID: 40015596 DOI: 10.1016/j.ejphar.2025.177430] [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: 10/28/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder worldwide, characterized by the loss of dopaminergic (DA) neurons in the substantia nigra and is associated with iron dyshomeostasis. Ferroptosis, a form of programmed cell death, involves iron-dependent lipid peroxidation and serves as a significant regulatory mechanism in PD. This study identified Tp53-induced glycolysis and apoptosis regulator (TIGAR) as a potential regulator of ferroptosis resistance in PD development. In this study, we demonstrated that in HT22 cells, 1-methyl-4-phenylpyridinium (MPP+) increased lipid peroxidation levels and reduced cell viability. These effects were reversed by the ferroptosis inhibitor ferrostatin-1 (Fer-1). MPP+ also induced elevated intracellular iron ion deposition, reactive oxygen species (ROS), and the lipid peroxidation product malondialdehyde (MDA). Meanwhile, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) significantly decreased glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH) levels, glutathione peroxidase (GPX) activity, and TIGAR expression, all of which were reversible with TIGAR overexpression. In an MPTP-induced in vivo PD model, TIGAR overexpression markedly increased DA neurons and reduced iron deposition. To summarize, TIGAR enhances intracellular NADPH production via the promotion of the pentose phosphate pathway (PPP), reduces intracellular glutathione disulfide (GSSG) to GSH, boosts GPX activity, and inhibits ferroptosis, thus providing neuronal protection.
Collapse
Affiliation(s)
- Yi-Chao Sheng
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China; Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Jia-Ni Huang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Wei-Long Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Rui Wan
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jing Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zheng-Hong Qin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yan Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
9
|
Wang H, Wang Q, Xu H, Wu Y, Cheung S, Xu Q, Pan C, Cao J, Cao Z, Yang R, Ding Y, Fei Y, Chen Y, Wang J, Liu C, Lu B. MEK1/2 inhibitors suppress pathological α-synuclein and neurotoxicity in cell models and a humanized mouse model of Parkinson's disease. Sci Transl Med 2025; 17:eadp4625. [PMID: 40367191 DOI: 10.1126/scitranslmed.adp4625] [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: 03/26/2024] [Revised: 11/30/2024] [Accepted: 03/04/2025] [Indexed: 05/16/2025]
Abstract
The abnormal accumulation of misfolded proteins is a common hallmark of many neurodegenerative disorders. Among these proteins, α-synuclein (αsyn) is a well-characterized pathogenic protein in Parkinson's disease (PD) and other synucleinopathies. αsyn can be hyperphosphorylated and form pathological aggregates, leading to neurodegeneration. Thus, chemical modulators of pathological αsyn may suppress its downstream toxicity and provide entry points to therapeutic intervention. Here, we identified mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitors as negative modulators of basal αsyn in wild-type cells and that pathological αsyn in αsyn preformed fibrils (αsyn-PFF) induced the neuroblastoma cell line SHSY-5Y, PC12 cells, and primary cultured neurons. We further demonstrated that these inhibitors suppressed Ser129 phosphorylated αsyn (p-αsyn) through the kinase PLK2 downstream of MEK1/2-ERK2 in PD cell models. We established a humanized PD mouse model by injecting human αsyn-PFF into mice with homozygous knock-in of human SNCA. Oral administration of blood-brain barrier-penetrable MEK1/2 inhibitors lowered pathological αsyn and rescued PD-relevant phenotypes with an acceptable safety profile in these mice. Collectively, these data highlight MEK1/2 inhibitors as a potential therapeutic strategy for PD.
Collapse
Affiliation(s)
- Huilan Wang
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Qing Wang
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Haoxiang Xu
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Yuanzheng Wu
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Siulam Cheung
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Qianhui Xu
- Interdisciplinary Research Center on Biology and Chemistry, State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Chengfang Pan
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jingyu Cao
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Zhiyuan Cao
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Ruonan Yang
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| | - Yu Ding
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yiyan Fei
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Yongfeng Chen
- Hangzhou Attec Biotech Co. Ltd., Room 406-409, Building 2, 1299 Zhangheng Road, Shanghai, China
| | - Jian Wang
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Shanghai Academy of Natural Sciences (SANS), Fudan University, Shanghai, China
| | - Boxun Lu
- Neurology Department at Huashan Hospital, School of Life Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
- New Cornerstone Science Laboratory, Fudan University, Shanghai 200438, China
| |
Collapse
|
10
|
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 DOI: 10.3390/cells14100684] [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: 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| |
Collapse
|
11
|
Intze A, Temperini ME, Rupert J, Polito R, Veber A, Puskar L, Schade U, Ortolani M, Zacco E, Tartaglia GG, Giliberti V. Effect of RNA on the supramolecular architecture of α-synuclein fibrils. Biophys J 2025:S0006-3495(25)00277-2. [PMID: 40329536 DOI: 10.1016/j.bpj.2025.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/20/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Structural changes associated with protein aggregation are challenging to study, requiring the combination of experimental techniques providing insights at the molecular level across diverse scales, ranging from nanometers to microns. Understanding these changes is even more complex when aggregation occurs in the presence of molecular cofactors such as nucleic acids and when the resulting aggregates are highly polymorphic. Infrared (IR) spectroscopy is a powerful tool for studying protein aggregates since it combines the label-free sensitivity to the cross-β architecture, an inherent feature of protein supramolecular aggregates, with the possibility to reach nanoscale sensitivity by leveraging atomic force microscopy (AFM)-assisted detection. Here, we present a combined approach that detects IR spectral markers of aggregation using various IR spectroscopy techniques, covering micro-to-nanoscale ranges, to study the effect of RNA on the supramolecular architecture of α-synuclein amyloid aggregates. We show a clear impact of RNA consistent with enhanced intermolecular forces, likely via a stronger hydrogen-bonded network stabilizing the cross-β architecture. AFM-assisted IR spectroscopy was crucial to assess that the more ordered the aggregates are, the stronger the structural impact of RNA. In addition, an RNA-induced reduction of the degree of polymorphism within the aggregate population is obtained.
Collapse
Affiliation(s)
- Antonia Intze
- Center for Life Nano- & Neuro-science, Istituto Italiano di Tecnologia (IIT), Rome, Italy; Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy; Department of Physics, Sapienza University of Rome, Rome, Italy.
| | - Maria Eleonora Temperini
- Center for Life Nano- & Neuro-science, Istituto Italiano di Tecnologia (IIT), Rome, Italy; Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Jakob Rupert
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy; Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy; Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Raffaella Polito
- Department of Physics, Sapienza University of Rome, Rome, Italy; Institute for Photonics and Nanotechnologies IFN-CNR, Rome, Italy
| | - Alexander Veber
- Institute for Electronic Structure Dynamics, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany; Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ljiljana Puskar
- Institute for Electronic Structure Dynamics, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany
| | - Ulrich Schade
- Institute for Electronic Structure Dynamics, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany
| | | | - Elsa Zacco
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Gian Gaetano Tartaglia
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy; Catalan Institution for Research and Advanced Studies, ICREA, Barcelona, Spain
| | - Valeria Giliberti
- Center for Life Nano- & Neuro-science, Istituto Italiano di Tecnologia (IIT), Rome, Italy.
| |
Collapse
|
12
|
Mather M. Autonomic dysfunction in neurodegenerative disease. Nat Rev Neurosci 2025; 26:276-292. [PMID: 40140684 DOI: 10.1038/s41583-025-00911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
In addition to their more studied cognitive and motor effects, neurodegenerative diseases are also associated with impairments in autonomic function - the regulation of involuntary physiological processes. These autonomic impairments manifest in different ways and at different stages depending on the specific disease. The neural networks responsible for autonomic regulation in the brain and body have characteristics that render them particularly susceptible to the prion-like spread of protein aggregation involved in neurodegenerative diseases. Specifically, the axons of these neurons - in both peripheral and central networks - are long and poorly myelinated axons, which make them preferential targets for pathological protein aggregation. Moreover, cortical regions integrating information about the internal state of the body are highly connected with other brain regions, which increases the likelihood of intersection with pathological pathways and prion-like spread of abnormal proteins. This leads to an autonomic 'signature' of dysfunction, characteristic of each neurodegenerative disease, that is linked to the affected networks and regions undergoing pathological aggregation.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Department of Psychology, University of Southern California, Los Angeles, CA, USA.
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Wang J, Geng T, Yao X, Liu Y. The relationship between serum GDF15 levels and non-motor symptoms in Parkinson's disease. Neurol Res 2025; 47:373-382. [PMID: 40099721 DOI: 10.1080/01616412.2025.2480331] [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: 11/26/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
OBJECTIVES The primary aim was to investigate the relationship between serum growth differentiation factor-15 (GDF15) levels and non-motor symptom (NMS) in Parkinson's disease (PD) patients. The secondary aim was to explore the diagnostic value of GDF15 for specific NMS. METHODS A total of 102 PD patients were enrolled in this study, including 47 males and 55 females. Doctors collected the clinical and demographic information of patients and detected the level of serum GDF15. Next, linear univariate and multivariate linear regression analyses were used to assess the correlation between GDF15 and NMS. Receiver operating characteristic curve analysis was performed to determine the optimal cut-off value of GDF15 and evaluate its diagnostic value. RESULTS In PD patients, there was no significant difference in serum GDF15 levels between males and females (p = 0.831). Age of PD onset, pesticide use, depression, sexual dysfunction, Epworth Sleepiness Scale (ESS) and Hamilton Depression Scale (HAMD) were associated with serum GDF15. Serum GDF15 was negatively correlated with HAMD, depression and sexual dysfunction and positively correlated with ESS. Each 10 pg/ml increase in serum GDF15 levels was associated with a 4% lower risk of depression and a 5% lower risk of sexual dysfunction. Notably, serum GDF15 may be a biomarker for distinguishing depression and sexual dysfunction in PD patients. CONCLUSION Elevated serum GDF15 reduced the risk of PD with depression and sexual dysfunction. Serum GDF15 may be a biomarker for distinguishing depression and sexual dysfunction in PD patients.
Collapse
Affiliation(s)
- Jianli Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
- Department of Geriatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ting Geng
- Department of Oncology, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Xiaomei Yao
- Department of Geriatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
14
|
Er S, Parkkinen I, Trepczyk K, Seelbach A, Pasculli MS, De Lorenzo F, Luk K, Jankowska E, Chmielarz P, Domanskyi A, Airavaara M. GDNF reduces fibril-induced early-stage alpha-synuclein pathology after delivery of 20S proteasome inhibitor lactacystin. Eur J Pharm Sci 2025; 208:107048. [PMID: 39988264 DOI: 10.1016/j.ejps.2025.107048] [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: 11/01/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Failures in protein homeostasis are linked to Parkinson's disease (PD) and other neurodegenerative diseases. Lewy bodies, proteinaceous inclusions rich in phosphorylated alpha-synuclein are a hallmark of PD. Glial cell line-derived neurotrophic factor (GDNF) can eliminate Lewy body-like inclusions in mouse dopamine neurons. This study explores whether GDNF has protective effects against alpha-synuclein protofibril toxicity under proteasome inhibition by lactacystin, both in vitro and in vivo. GDNF did not shield midbrain dopamine neurons from lactacystin-induced neurodegeneration, but still prevented phosphorylated alpha-synuclein accumulation. In vivo experiment with control or GDNF-expressing viral vectors assessed alpha-synuclein pathology spread in the nigrostriatal pathway and lactacystin-caused damage in the midbrain. GDNF overexpression reduced phosphorylated alpha-synuclein inclusions. Lactacystin-treated mice showed motor asymmetry and decreased spontaneous activity, exacerbated without AAV-GDNF pre-treatment. However, GDNF's neuroprotective effect could not be confirmed in vivo, due to side-effects from overexpression in the midbrain. Importantly, these findings show that GDNF continues to eliminate alpha-synuclein aggregation despite lactacystin-induced proteasome inhibition. Activating neurotrophic signaling pathways may protect against alpha-synuclein pathology in PD, even with impaired protein degradation mechanisms.
Collapse
Affiliation(s)
- Safak Er
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Ilmari Parkkinen
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Karolina Trepczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Anna Seelbach
- Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | | | - Francesca De Lorenzo
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elzbieta Jankowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Mikko Airavaara
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
15
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [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: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
16
|
Huang Z, Pan Y, Ma K, Luo H, Zong Q, Wu Z, Zhu Z, Guan Y. Nicotine Ameliorates α-Synuclein Preformed Fibril-Induced Behavioral Deficits and Pathological Features in Mice. Appl Biochem Biotechnol 2025; 197:3026-3047. [PMID: 39815141 DOI: 10.1007/s12010-024-05086-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] [Accepted: 11/12/2024] [Indexed: 01/18/2025]
Abstract
Epidemiologic study suggests that nicotine reduces the risk of Parkinson's disease (PD) and thus could serve as a potential treatment. In this study, we aimed to investigate the effect of nicotine on the behavioral phenotypes and pathological characteristics of mice induced by human alpha-synuclein preformed fibers (α-syn-PFF). Mice were injected with 5 µg of human α-syn-PFF in the hippocampus while administering nicotine-containing drinking water (200 µg/mL). After 1 month, the motor ability, mood, spatial learning, and memory ability of the PD phenotype-like model mice were detected using open field, rotarod, Y maze, and O maze tests. The expression of pathological α-syn and apoptotic proteins, as well as the number of glial and neural stem cells in the hippocampus of mice, was detected using western blot and immunofluorescence. The results demonstrated that nicotine significantly reduced pathological α-syn accumulation, α-syn serine 129 phosphorylation, and apoptosis induced by α-syn-PFF injection in the hippocampus of mice. Nicotine also inhibited the increase in the number of glia, microglia, and neuronal apoptotic cells, and it decreased the expression of PI3K and Akt while also exhibiting significant memory impairment, motor deficits, and anxiety-like behavior. In conclusion, our findings suggest that nicotine ameliorates behavioral deficits and pathological changes in mice by inhibiting human α-syn-PFF-induced neuroinflammation and apoptosis.
Collapse
Affiliation(s)
- Zhangqiong Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Kaili Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Haiyu Luo
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Qinglan Zong
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Zhengcun Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 935, Jiaoling Road, Kunming, 650118, China
| | - Zhouhai Zhu
- The Joint Institute of Tobacco and Health, No. 367, Honglin Road, Kunming, 650231, China.
| | - Ying Guan
- The Joint Institute of Tobacco and Health, No. 367, Honglin Road, Kunming, 650231, China.
| |
Collapse
|
17
|
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
|
18
|
Diao XJ, Soto C, Wang F, Wang Y, Wu YC, Mukherjee A. The potential of brain organoids in addressing the heterogeneity of synucleinopathies. Cell Mol Life Sci 2025; 82:188. [PMID: 40293500 PMCID: PMC12037466 DOI: 10.1007/s00018-025-05686-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: 09/16/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Synucleinopathies are a group of diseases characterized by neuronal and glial accumulation of α-synuclein (aSyn) linked with different clinical presentations, including Parkinson's disease (PD), Parkinson's disease with dementia (PDD), Dementia with Lewy Bodies (DLB) and Multiple system atrophy (MSA). Interestingly, the structure of the aSyn aggregates can vary across different synucleinopathies. Currently, it is unclear how the aSyn protein can aggregate into diverse structures and affect distinct cell types and various brain regions, leading to different clinical symptoms. Recent advances in induced pluripotent stem cells (iPSCs)-based brain organoids (BOs) technology provide an unprecedented opportunity to define the etiology of synucleinopathies in human brain cells within their three-dimensional (3D) context. In this review, we will summarize current advances in investigating the mechanisms of synucleinopathies using BOs and discuss the scope of this platform to define mechanisms underlining the selective vulnerability of cell types and brain regions in synucleinopathies.
Collapse
Affiliation(s)
- Xiao-Jun Diao
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fei Wang
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Abhisek Mukherjee
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
19
|
Zhou N, Chen J, Hu M, Wen N, Cai W, Li P, Zhao L, Meng Y, Zhao D, Yang X, Liu S, Huang F, Zhao C, Feng X, Jiang Z, Xie E, Pan H, Cen Z, Chen X, Luo W, Tang B, Min J, Wang F, Yang J, Xu H. SLC7A11 is an unconventional H + transporter in lysosomes. Cell 2025:S0092-8674(25)00406-4. [PMID: 40280132 DOI: 10.1016/j.cell.2025.04.004] [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: 04/14/2024] [Revised: 01/22/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Lysosomes maintain an acidic pH of 4.5-5.0, optimal for macromolecular degradation. Whereas proton influx is produced by a V-type H+ ATPase, proton efflux is mediated by a fast H+ leak through TMEM175 channels, as well as an unidentified slow pathway. A candidate screen on an orphan lysosome membrane protein (OLMP) library enabled us to discover that SLC7A11, the protein target of the ferroptosis-inducing compound erastin, mediates a slow lysosomal H+ leak through downward flux of cystine and glutamate, two H+ equivalents with uniquely large but opposite concentration gradients across lysosomal membranes. SLC7A11 deficiency or inhibition caused lysosomal over-acidification, reduced degradation, accumulation of storage materials, and ferroptosis, as well as facilitated α-synuclein aggregation in neurons. Correction of abnormal lysosomal acidity restored lysosome homeostasis and prevented ferroptosis. These studies have revealed an unconventional H+ transport conduit that is integral to lysosomal flux of protonatable metabolites to regulate lysosome function, ferroptosis, and Parkinson's disease (PD) pathology.
Collapse
Affiliation(s)
- Nan Zhou
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Jingzhi Chen
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Meiqin Hu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China.
| | - Na Wen
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Weijie Cai
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Liding Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Meng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Dongdong Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaotong Yang
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Siyu Liu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Fangqian Huang
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Xinghua Feng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Zikai Jiang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Enjun Xie
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongxu Pan
- Department of Neurology & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhidong Cen
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhui Chen
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Luo
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beisha Tang
- Department of Neurology & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junxia Min
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Junsheng Yang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Haoxing Xu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research and The State Key Lab of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
20
|
Takahashi R, Yamakado H, Uemura N, Taguchi T, Ueda J. The Gut-Brain Axis Based on α-Synuclein Propagation-Clinical, Neuropathological, and Experimental Evidence. Int J Mol Sci 2025; 26:3994. [PMID: 40362234 PMCID: PMC12072079 DOI: 10.3390/ijms26093994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
The cytopathological hallmark of Parkinson's disease (PD) is a neuronal cytoplasmic inclusion called Lewy body (LB). Lewy bodies are composed of alpha-synuclein (aSyn), a 140 aa protein that is predominantly expressed in the presynaptic terminal and which is implicated in neurotransmitter release. Recently, aSyn was found to propagate from neuron to neuron in a trans-synaptic manner. Although the precise molecular mechanisms are unclear, the propagation of aSyn is believed to play a major role in the progression of Lewy pathology in PD. Neuropathologically, the initial Lewy pathology has been shown to be formed in the dorsal motor nucleus of the vagus (DMV) or olfactory bulb by neuropathological studies. Since the DMV innervates the enteric nervous system (ENS) and LBs are formed in the gut nerve plexuses, it is conceivable that LBs propagate from the gut to the DMV and then to other regions of the brain. In this article, clinical, neuropathological, and experimental evidence supporting or negating the idea that aSyn propagation from the ENS to the brain leads to PD is reviewed. Moreover, the propagation of aSyn seeds through systemic circulation or multifocal generation of aSyn seeds is discussed as a potential alternative scenario for aSyn spreading.
Collapse
Affiliation(s)
- Ryosuke Takahashi
- Kyoto University Office of Research Acceleration, Kyoto 606-8501, Japan
| | - Hodaka Yamakado
- Department of Therapeutics for Multiple System Atrophy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan;
| | - Norihito Uemura
- Department of Neurological Disease Control, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Tomoyuki Taguchi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (T.T.); (J.U.)
| | - Jun Ueda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (T.T.); (J.U.)
| |
Collapse
|
21
|
Boschen SL, A Mukerjee A, H Faroqi A, E Rabichow B, Fryer J. Research models to study lewy body dementia. Mol Neurodegener 2025; 20:46. [PMID: 40269912 PMCID: PMC12020038 DOI: 10.1186/s13024-025-00837-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: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Lewy body dementia (LBD) encompasses neurodegenerative dementias characterized by cognitive fluctuations, visual hallucinations, and parkinsonism. Clinical differentiation of LBD from Alzheimer's disease (AD) remains complex due to symptom overlap, yet approximately 25% of dementia cases are diagnosed as LBD postmortem, primarily identified by the presence of α-synuclein aggregates, tau tangles, and amyloid plaques. These pathological features position LBD as a comorbid condition of both Parkinson's disease (PD) and AD, with over 50% of LBD cases exhibiting co-pathologies. LBD's mixed pathology complicates the development of comprehensive models that reflect the full spectrum of LBD's etiological, clinical, and pathological features. While existing animal and cellular models have facilitated significant discoveries in PD and AD research, they lack specificity in capturing LBD's unique pathogenic mechanisms, limiting the exploration of therapeutic avenues for LBD specifically. This review assesses widely used PD and AD models in terms of their relevance to LBD, particularly focusing on their ability to replicate human disease pathology and assess treatment efficacy. Furthermore, we discuss potential modifications to these models to advance the understanding of LBD mechanisms and propose innovative research directions aimed at developing models with enhanced etiological, face, predictive, and construct validity.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
- Department of Neurosurgery, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Aarushi A Mukerjee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ayman H Faroqi
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ben E Rabichow
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - John Fryer
- Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 850054, USA
| |
Collapse
|
22
|
Lai TT, Xiang W, Stanojlovic M, Käufer C, Feja M, Lau K, Zunke F, Richter F. The basolateral amygdala and striatum propagate alpha-synuclein pathology causing increased fear response in a Parkinson's disease model. Brain Behav Immun 2025; 128:469-486. [PMID: 40274000 DOI: 10.1016/j.bbi.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/30/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Alpha-synuclein (aSyn)-related pathology crucially contributes to the pathogenesis of Parkinson's disease, a frequent and incurable neurodegenerative disease characterized by progressive motor and non-motor symptoms. Anxiety and fear- related neuropsychiatric symptoms develop frequently and early in the disease, but a lack of understanding of pathogenesis hampers rational therapy. This study aimed to decipher whether aSyn pathology in the basolateral amygdala (BLA) is causative of fear and anxiety. Bilateral stereotaxic injections of human aSyn-preformed amyloid fibrils (PFF) in BLA, striatum, or substantia nigra were conducted in female mice overexpressing human aSyn (Thy1-aSyn) and in wildtype littermates (WT). We characterized the propagation of aSyn pathology and related neuropathological changes across brain regions and examined the behavioral and fear responses in mice up to 2 months post-injection of PFF. While PFF injections induced local aSyn fibril pathology close to all respective injection sites in transgenic mice, we observed differences in propagation, downstream pathology and behavioral alterations. The BLA and the striatum, but not the substantia nigra, effectively propagated aSyn pathology to connected brain regions at 2.5 months post injection. This involved enhanced microgliosis and astrogliosis in the nigrostriatal system and loss of GABAergic parvalbuminergic interneurons in the striatum and corticolimbic brain regions. Intra-BLA PFF injections resulted in increased cued fear response in both transgenic mice and WT mice at 1 month post injection. The effect was more pronounced in the transgenic mice. Conversely, intra-striatal PFF injections enhanced contextual fear in WT at 2 months post injection. These findings imply that increased fear is inducible by aSyn pathology, especially if originating in the BLA or striatum. Furthermore, both regions are hub regions of aSyn pathology propagation, thereby contributing to disease progression. These insights provide mechanisms that can guide rational therapeutic development.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Milos Stanojlovic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Christopher Käufer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| |
Collapse
|
23
|
Vroman R, de Lichtervelde L, Singh Dolt K, Robertson G, Kriek M, Barbato M, Cholewa-Waclaw J, Kunath T, Downey P, Zagnoni M. A high-fidelity microfluidic platform reveals retrograde propagation as the main mechanism of α-Synuclein spread in human neurons. NPJ Parkinsons Dis 2025; 11:80. [PMID: 40254612 PMCID: PMC12009960 DOI: 10.1038/s41531-025-00936-x] [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: 11/21/2024] [Accepted: 04/05/2025] [Indexed: 04/22/2025] Open
Abstract
α-Synuclein (αSyn) is a major component of Lewy bodies and Lewy neurites, which are a pathological hallmark of Parkinson's disease (PD). Pathologically aggregated forms of αSyn can spread along neurites and induce the misfolding of normal αSyn. To elucidate how αSyn pathology propagates between brain areas, we developed a novel in vitro microfluidic platform to study the intracellular transport of preformed fibrils and the induction and spread of αSyn aggregates. Patient-derived midbrain dopaminergic (mDA) neurons were cultured in microfluidic devices designed to maintain unidirectional axonal connections between fluidically isolated mDA neuronal cultures for over 3 months. Using αSyn preformed fibrils to induce Lewy-like pathology, we found that anterograde spread of αSyn fibrils was slow and occurred at low levels, while retrograde spread was significantly more efficient. This is in line with observations in animal models and shows that the platform provides an innovative new tool for studying PD in vitro.
Collapse
Affiliation(s)
- Rozan Vroman
- Center for Microsystems and Photonics, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | | | - Karamjit Singh Dolt
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Graham Robertson
- Center for Microsystems and Photonics, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK
| | | | - Michela Barbato
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Justyna Cholewa-Waclaw
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Patrick Downey
- UCB Biopharma, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium
| | - Michele Zagnoni
- Center for Microsystems and Photonics, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
24
|
Hage A, Janes M, Best SM. A No-Brainer! The Therapeutic Potential of TRIM Proteins in Viral and Central Nervous System Diseases. Viruses 2025; 17:562. [PMID: 40285004 PMCID: PMC12031127 DOI: 10.3390/v17040562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Tripartite motif (TRIM) proteins comprise an important class of E3 ubiquitin ligases that regulate numerous biological processes including protein expression, cellular signaling pathways, and innate immunity. This ubiquitous participation in fundamental aspects of biology has made TRIM proteins a focus of study in many fields and has illuminated the negative impact they exert when functioning improperly. Disruption of TRIM function has been linked to the success of various pathogens and separately to the occurrence and development of several neurodegenerative diseases, making TRIM proteins an appealing candidate to study for novel therapeutic approaches. Here, we review the current findings on TRIM proteins that demonstrate their analogous properties in the distinct fields of viral infection and central nervous system (CNS) disorders. We also examine recent advancements in drug development and targeted protein degradation as potential strategies for TRIM-mediated therapeutic treatments and discuss the implications these technologies have on future research directions.
Collapse
Affiliation(s)
- Adam Hage
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA; (M.J.); (S.M.B.)
| | | | | |
Collapse
|
25
|
Zhao X, Cao R, Tian X, Liu P, Liu D, Yu X, Zheng Z, Chen GL, Zou L. OAB-14 Attenuated Glymphatic System Disorder, Neuroinflammation and Dyskinesia in Parkinson's Disease Model Mice Induced by Rotenone. Neurochem Res 2025; 50:142. [PMID: 40220255 DOI: 10.1007/s11064-025-04388-w] [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: 01/27/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder characterized by the pathological accumulation of alpha-synuclein (α-syn) in the neuronal cell bodies of the substantia nigra. The glymphatic system within the Central Nervous System (CNS) is responsible for clearing metabolic waste and abnormal proteins and its dysfunction may significantly contribute to the pathogenesis of PD. Our previous study showed that OAB-14, the novel small molecular compound, showed a great potential effect in APP/PS1 transgenic mice. Given the similarities in the pathogenesis of PD and Alzheimer's disease (AD), it is pertinent to explore the therapeutic potential of OAB-14 in the context of PD. This study utilized a rotenone-induced PD mice model to evaluate the effects of oral administration of OAB-14, and its underlying mechanisms. Here we confirmed the neuroprotective effect and motor improvement of OAB-14 in rotenone-induced PD model mice. Our research has shown that OAB-14 is capable of enhancing the glymphatic system function by promoting the influx and efflux of the CSF tracers to the brain and deep cervical lymph nodes, respectively, to promote the clearance of α-syn. In addition, OAB-14 could down-regulate MyD88, NF-kB (Ser 536) phosphorylation, and TLR4 to reduce glial cell activation; and down-regulate cleaved-caspase1, NLRP3, ASC, IL-1β, IL-6, IL-18, TNF-α, and IL-10 to reduce the expression of inflammatory vesicles and pro-inflammatory factors, and to reduce neuronal oxidative stress. In summary, OAB-14 may promote the clearance of brain α-syn through the glial lymphatic system, inhibit the α-syn/TLR4/NF-κB/NLRP3 inflammatory pathway, and improve movement disorders.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Ruolin Cao
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Xiaoyi Tian
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Peng Liu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Danyang Liu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Xin Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| | - Zhonghui Zheng
- Shandong Xinhua Pharmaceutical Co., Ltd, Zibo, Shandong, 255086, PR China
| | - Guo-Liang Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| | - Libo Zou
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
26
|
Mishra R, Upadhyay A. An update on mammalian and non-mammalian animal models for biomarker development in neurodegenerative disorders. Cell Mol Life Sci 2025; 82:147. [PMID: 40192808 PMCID: PMC11977071 DOI: 10.1007/s00018-025-05668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Neurodegeneration is one of the leading factor for death globally, affecting millions of people. Developing animal models are critical to understand biological processes and comprehend pathological hallmarks of neurodegenerative diseases. For decades, many animal models have served as excellent tools to determine the disease progression, develop diagnostic methods and design novel therapies against distinct pathologies. Here, we provide a comprehensive overview of both, mammalian and non-mammalian animal models, with a focus on three most common and aggressive neurodegenerative disorders: Alzheimer's disease, Parkinson's disease and Spinocerebellar ataxia-1. We highlight various approaches including transgene, gene transfer, and chemically-induced methods used to develop disease models. In particular, we discuss applications of both non-mammalian and mammalian contributions in research on neurodegeneration. It is exciting to learn the roles of animal models in disease pathomechanisms, identifying biomarkers and hence devising novel interventions to treat neuropathological conditions.
Collapse
Affiliation(s)
- Ribhav Mishra
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, 491002, India
| |
Collapse
|
27
|
Quansah E, Vatsa N, Ensink E, Brown J, Cave T, Aguileta M, Schulz E, Lindquist A, Gilliland C, Steiner JA, Escobar Galvis ML, Milčiūtė M, Henderson MX, Brundin P, Brundin L, Marshall LL, Gordevicius J. Tet2 loss and enhanced ciliogenesis suppress α-synuclein pathology. Acta Neuropathol Commun 2025; 13:71. [PMID: 40189544 PMCID: PMC11974201 DOI: 10.1186/s40478-025-01988-z] [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: 10/26/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
There are no approved treatments that slow Parkinson's disease (PD) progression and therefore it is important to identify novel pathogenic mechanisms that can be targeted. Loss of the epigenetic marker, Tet2 appears to have some beneficial effects in PD models, but the underlying mechanism of action is not well understood. We performed an unbiased transcriptomic analysis of cortical neurons isolated from patients with PD to identify dysregulated pathways and determine their potential contributions to the disease process. We discovered that genes associated with primary cilia, non-synaptic sensory and signaling organelles, are upregulated in both early and late stage PD patients. Enhancing ciliogenesis in primary cortical neurons via sonic hedgehog signaling suppressed the accumulation of α-synuclein pathology in vitro. Interestingly, deletion of Tet2 in mice also enhanced the expression of primary cilia and sonic hedgehog signaling genes and reduced the accumulation of α-synuclein pathology and dopamine neuron degeneration in vivo. Our findings demonstrate the crucial role of TET2 loss in regulating ciliogenesis and potentially affecting the progression of PD pathology.
Collapse
Affiliation(s)
- Emmanuel Quansah
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA.
| | - Naman Vatsa
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Elizabeth Ensink
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jaycie Brown
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Tyce Cave
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Miguel Aguileta
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Emily Schulz
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Allison Lindquist
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Carla Gilliland
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jennifer A Steiner
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | | | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Patrik Brundin
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Roche Pharma Research and Early Development (pRED), Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, Basel, Switzerland
| | - Lena Brundin
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | | |
Collapse
|
28
|
Landolfi A, Sorrentino C, Barone P, Erro R. Biological frameworks for Parkinson's disease: the heterogeneity SAAgged. J Neurol 2025; 272:318. [PMID: 40186646 DOI: 10.1007/s00415-025-13049-5] [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: 02/03/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Recent biological frameworks of Parkinson's disease (PD) rely on the new advances in α-synuclein detection in biological tissues, mostly through α-synuclein seed amplification assays, and are mainly aimed at intercepting pre-clinical or early phases of disease to be subjected to disease-modifying therapies targeting α-synuclein. However, α-synuclein pathology alone is insufficient to explain the observed clinical heterogeneity of PD. Indeed, it has been demonstrated that a number of additional elements, such as genetics, comorbidities, co-pathology, and environmental factors, may influence PD phenotype and progression. Such factors have been partially accounted for or completely overlooked by both biological frameworks and would instead represent features which could explain, at least partially, the clinical and pathophysiologic diversities of PD and further represent potential druggable targets. Recognizing that the clinical heterogeneity of PD is a window to understand the pathophysiologic complexity of the disease might turn useful for a refinement of the current biological frameworks and move the field to satisfy the unmet need of establishing a precision medicine framework for this prevalent disorder.
Collapse
Affiliation(s)
- Annamaria Landolfi
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, SA, Italy
| | - Cristiano Sorrentino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, SA, Italy
| | - Paolo Barone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, SA, Italy
- IRCCS Synlab SDN, Naples, Italy
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081, Baronissi, SA, Italy.
| |
Collapse
|
29
|
Zhan A, Zhong K, Zhang K. Novel subcellular regulatory mechanisms of protein homeostasis and its implications in amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2025; 756:151582. [PMID: 40056503 DOI: 10.1016/j.bbrc.2025.151582] [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: 12/12/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron degenerative disorder. Protein aggregates induce various forms of neuronal dysfunction and represent pathological hallmarks in ALS patients. Reducing protein aggregates could be a promising therapeutic strategy for ALS. While most studies have focused on cytoplasmic protein homeostasis, neurons adaptively reduce aggregates across subcellular compartments during stress through previously uncharacterized mechanisms. Here, we summarize novel compartment-specific proteostatic mechanisms: (1) the ERAD/RESET pathways, (2) HSPs-mediated nuclear sequestration, (3) mitochondrial aggregate import (MAGIC), (4) neurite-localized UPS/autophagosome and NMP, and (5) exopher-mediated extracellular disposal. These mechanisms collectively ensure cellular stress adaptation and provide novel therapeutic targets for ALS treatment.
Collapse
Affiliation(s)
- Aisheng Zhan
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Keke Zhong
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Kejing Zhang
- Institute of Translational Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| |
Collapse
|
30
|
Liang Z, Murugappan SK, Li Y, Lai MN, Qi Y, Wang Y, Chan HYE, Lee MM, Chan MK. Gene delivery of SUMO1-derived peptide rescues neuronal degeneration and motor deficits in a mouse model of Parkinson's disease. Mol Ther 2025:S1525-0016(25)00279-5. [PMID: 40189878 DOI: 10.1016/j.ymthe.2025.04.005] [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: 07/23/2024] [Revised: 12/18/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Developing α-synuclein aggregation inhibitors is challenging because its aggregation process involves several microscopic steps and heterogeneous intermediates. Previously, we identified a SUMO1-derived peptide, SUMO1(15-55), that exhibits tight binding to monomeric α-synuclein via SUMO-SUMO-interacting motif (SIM) interactions, and effectively blocks the initiation of aggregation and formation of toxic aggregates in vitro. In cellular and Drosophila models, SUMO1(15-55) was efficacious in protecting neuronal cells from α-synuclein-induced neurotoxicity and neuronal degeneration. Given the demonstrated ability of SUMO1(15-55) to sequester α-synuclein monomers thereby blocking oligomer formation, we sought to evaluate whether it could be equally effective against the aggregation-prone familial mutant α-synuclein-A53T. Herein, we show that SUMO1(15-55) selectively binds to monomeric α-synuclein-A53T, inhibits primary nucleation, and prevents the formation of structured protofibrils in vitro, thereby protecting neuronal cells from protofibril-induced cell death. We further demonstrate that larval feeding of a designed His10-SUMO1(15-55) that exhibits enhanced sub-stoichiometric suppression of α-synuclein-A53T aggregation in vitro can ameliorate Parkinson's disease (PD)-related symptoms in α-synuclein-A53T transgenic Drosophila models, while its rAAV-mediated gene delivery can relieve the PD-related histological and behavioral deficiencies in an rAAV-α-synuclein-A53T mouse PD model. Our findings suggest that gene delivery of His10-SUMO1(15-55) may serve as a clinical therapy for a spectrum of α-synuclein-aggregation associated synucleinopathies.
Collapse
Affiliation(s)
- Zhaohui Liang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Suresh Kanna Murugappan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Yuxuan Li
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Man Nga Lai
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Yajing Qi
- Department of Physics, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Yi Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Ho Yin Edwin Chan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Laboratory of Drosophila Research, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - Marianne M Lee
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China.
| | - Michael K Chan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China; Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin 999077, Hong Kong SAR, China.
| |
Collapse
|
31
|
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
|
32
|
Phan L, Miller D, Gopinath A, Lin M, Gunther D, Kiel K, Quintin S, Borg D, Hasanpour-Segherlou Z, Newman A, Sorrentino Z, Miller E J, Seibold J, Hoh B, Giasson B, Khoshbouei H. Parkinson's Paradox: Alpha-synuclein's Selective Strike on SNc Dopamine Neurons over VTA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644952. [PMID: 40236072 PMCID: PMC11996431 DOI: 10.1101/2025.03.24.644952] [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
In synucleinopathies, including Parkinson's disease (PD), dopamine neurons in the substantia nigra pars compacta (SNc) exhibit greater vulnerability to degeneration than those in the ventral tegmental area (VTA). While α-synuclein (αSyn) pathology is implicated in nigral dopamine neuron loss, the mechanisms by which αSyn affects neuronal activity and midbrain dopamine network connectivity prior to cell death remain unclear. This study tested the hypothesis that elevated αSyn expression induces pathophysiological changes in firing activity and disrupts network connectivity dynamics of dopamine neurons before neuronal loss. We employed two mouse models of synucleinopathy: preformed αSyn fibril (PFF) injection and AAV-mediated expression of human αSyn (hαSyn) under the control of the tyrosine hydroxylase (TH) promoter, both targeting the VTA and SNc. Four weeks post-injection, brain sections underwent histological, electrophysiological, and network analyses. Immunohistochemistry for TH, hαSyn, and phospho-Ser129 αSyn assessed αSyn expression and dopaminergic neuron alterations. Neuronal viability was evaluated using two complementary approaches: quantification of TH + or FOX3 + and TUNEL labeling. Importantly, these analyses revealed no significant changes in neuronal counts or TUNEL + cells at this time point, confirming that subsequent functional assessments captured pre-neurodegenerative, αSyn-induced alterations rather than late-stage neurodegeneration. Electrophysiological recordings revealed a differential effect of hαSyn expression. SNc dopamine neurons exhibited significantly increased baseline firing rates, whereas VTA dopamine neurons remained unchanged. These findings indicate a region-specific vulnerability to αSyn-induced hyperactivity of dopamine neurons. Further analysis revealed impaired homeostatic firing rate regulation in SNc, but not VTA, dopamine neurons, demonstrated by a reduced capacity to recover baseline firing following hyperpolarization. Collectively, our results demonstrate that, prior to neurodegeneration, elevated αSyn expression differentially disrupts both basal firing activity and network stability of SNc dopamine neurons, while sparing VTA dopamine neurons. By identifying neurophysiological changes preceding dopaminergic neuron loss, these findings provide critical insights into the pathophysiological mechanisms predisposing SNc neurons to degeneration in Parkinson's disease. Significance Statement A central question in Parkinson's disease research is why dopamine neurons in the substantia nigra pars compacta (SNc) are more vulnerable than those in the ventral tegmental area (VTA). This study reveals that alpha-synuclein (αSyn) pathology differentially impacts dopamine neuronal activity and network connectivity, causing changes in the SNc before neuronal loss occurs, but not in the VTA. These findings provide a mechanism to explain the differential resilience of these neighboring dopamine neuron populations and provide insights into Parkinson's disease progression. The methodologies developed in this study establish a foundation for investigating network topology in deep brain structures and its role in neurodegenerative disorders.
Collapse
|
33
|
Fleming SM, Scott S, Hamad EJ, Herman DE, Holden JG, Yan L, Linning-Duffy K, Kemp CJ, Patterson JR, Miller KM, Kubik M, Kuhn N, Stoll AC, Duffy MF, Steece-Collier K, Cole-Strauss A, Lipton JW, Luk KC, Sortwell CE. Intrastriatal injection of alpha-synuclein preformed fibrils to rats results in L-DOPA reversible sensorimotor impairments and alterations in non-motor function. Front Neurosci 2025; 19:1556447. [PMID: 40236948 PMCID: PMC11996896 DOI: 10.3389/fnins.2025.1556447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction The alpha-synuclein (α-syn) preformed fibril (PFF) model of Parkinson's disease (PD) is widely used in rodents to understand the mechanisms contributing to progression of pathology and neurodegeneration in the disorder. While the time course of pathology in the α-syn PFF rat model has been well characterized, it has been more challenging to determine reliable and reproducible behavior impairments. This is mainly due to α-syn PFF injections resulting in a partial nigrostriatal lesion that make motor anomalies more subtle and difficult to detect, just as in patients with PD. In the present study we sought to examine the effect of increased striatal distribution and injection quantity of α-syn PFFs in rats on accumulation of phosphorylated α-syn inclusions, nigrostriatal degeneration, sensorimotor behavior, and nonmotor function related to PD. Methods Male Fischer 344 rats were injected unilaterally in the striatum with a total of 24μg α-syn PFFs distributed into three sites, or an equal volume of phosphate buffered saline (PBS) as a control condition. Sensorimotor function was assessed using a battery of behavioral tests sensitive to varying degrees of nigrostriatal neurodegeneration. Non-motor testing included assays for olfaction, emotional reactivity, cognitive function, and sleep. Results At six months post injection, α-syn PFF rats displayed significant movement and somatosensory asymmetries compared with control rats. Time to initiate a forelimb step and time to contact an adhesive stimulus on the forepaw took significantly longer with the contralateral limb compared with the ipsilateral limb in α-syn PFF rats. Further, hindlimb stepping in the cylinder was significantly reduced in α-syn PFF-injected rats compared with controls. Cognitive function was also affected in the α-syn PFF rats, with investigation time significantly decreased in an object recognition test. Levodopa reversibility was observed in the movement initiation and cylinder tests. Postmortem analysis revealed a 55% loss of nigral tyrosine hydroxylase immunoreactive neurons and a 63% reduction in striatal dopamine content in α-syn PFF-injected rats. Conclusion Thus, using the present α-syn PFF surgical parameters, sufficient nigrostriatal degeneration can be achieved to manifest significant motor and non-motor deficits. These rat α-syn PFF surgical parameters will be important for preclinical assessment of novel diseasemodifying therapies.
Collapse
Affiliation(s)
- Sheila M. Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sophia Scott
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Edward J. Hamad
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Danielle E. Herman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - John G. Holden
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Christopher J. Kemp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Joseph R. Patterson
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathryn M. Miller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Nathan Kuhn
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Anna C. Stoll
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Megan F. Duffy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Jack W. Lipton
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kelvin C. Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Caryl E. Sortwell
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| |
Collapse
|
34
|
Zhao L, Cao Y, Xin Y, Liu C, Yang J, Li Y, Tian S, Liu Z, Jia H, Liu M, Hu M, Luo L, Meng F. Targeted Raman Visualization and Mitigation of α-Synuclein Amyloidogenesis in Living Zebrafish by a Nanobody-Decorated Polydiacetylene. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411419. [PMID: 39996265 DOI: 10.1002/smll.202411419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/03/2025] [Indexed: 02/26/2025]
Abstract
α-Synuclein (α-Syn) amyloidogenesis is considered a promising diagnostic marker and therapeutic target for Parkinson's disease (PD). Simultaneously visualizing and mitigating α-Syn amyloidogenesis are essential for future PD theranostics, yet they continue to pose an insurmountable challenge. This study have herein developed a nanobody-decorated polydiacetylene to approach a straightforward solution. Grafting α-Syn61-95 segment into the third complementary determining region of a parent nanobody generates an engineered nanobody X30 that can bind with α-Syn and prevent its amyloidogenesis through homotypic interaction. It next use X30 to decorate poly(deca-4,6-diynedioic acid) (PDDA), a polydiacetylene with an ultrastrong alkyne Raman signal (2120 cm-1) in the cellular silent region, to create an α-Syn targeting Raman probe PX30. The binding affinity between X30 and α-Syn can be further boosted for over 150 times attributed to the rigidity of PDDA backbone and the multivalent effect. Therefore, PX30 not only enables real-time Raman visualization of α-Syn amyloidogenesis with a high signal-to-noise ratio in living zebrafish, but also alleviates amyloidogenesis-mediated damage to zebrafish embryos by effectively inhibiting α-Syn amyloidogenesis at low stoichiometric concentrations and scavenging pathologic reactive oxygen species.
Collapse
Affiliation(s)
- Liyuan Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yujuan Cao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yanru Xin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Chenxi Liu
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Jin Yang
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Yanan Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zhenxing Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Min Hu
- Hubei Institute for Drug Control, Wuhan, 430075, P. R. China
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry, Materia Medica School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry, Materia Medica School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
35
|
Li X, Bi L, Zhang S, Xu Q, Xia W, Tao Y, Wu S, Li Y, Le W, Kang W, Li D, Sun B, Liu C. Single-Molecule Insight Into α-Synuclein Fibril Structure and Mechanics Modulated by Chemical Compounds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416721. [PMID: 39951335 PMCID: PMC11984887 DOI: 10.1002/advs.202416721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Indexed: 04/12/2025]
Abstract
α-Syn fibrils, a key pathological hallmark of Parkinson's disease, is closely associated with disease initiation and progression. Several small molecules are found to bind or dissolve α-syn fibrils, offering potential therapeutic applications. Here, an innovative optical tweezers-based, fluorescence-combined approach is developed to probe the mechanical characteristics of α-syn fibrils at the single-molecule level. When subjected to axial stretching, local deformation within α-syn fibrils appeared at forces above 50 pN. These structural alternations occurred stepwise and are irreversible, suggesting unfolding of individual α-syn molecules or subdomains. Additionally, α-syn fibrils exhibits high heterogeneity in lateral disruption, with rupture force ranging from 50 to 500 pN. The impact of different compounds on the structure and mechanical features of α-syn fibrils is further examined. Notably, epigallocatechin gallate (EGCG) generally attenuates the rupture force of fibrils by wedging into the N-terminal polar groove and induces fibril dissociation. Conversely, copper chlorophyllin A (CCA) attaches to four different sites wrapping around the fibril core, reinforcing the stability of the fibril against rupture forces. The work offers an effective method for characterizing single-fibril properties and bridges compound-induced structural alternations with mechanical response. These insights are valuable for understanding amyloid fibril mechanics and their regulation by small molecules.
Collapse
Affiliation(s)
- Xiang Li
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200030China
- Zhangjiang Institute for Advanced StudyShanghai Jiao Tong UniversityShanghai201203China
| | - Lulu Bi
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Shenqing Zhang
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200030China
- Zhangjiang Institute for Advanced StudyShanghai Jiao Tong UniversityShanghai201203China
| | - Qianhui Xu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai201210China
- University of the Chinese Academy of SciencesChinese Academy of SciencesBeijing100049China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai201210China
- University of the Chinese Academy of SciencesChinese Academy of SciencesBeijing100049China
| | - Youqi Tao
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200030China
- Zhangjiang Institute for Advanced StudyShanghai Jiao Tong UniversityShanghai201203China
| | - Shaojuan Wu
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Yanan Li
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Weidong Le
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu HospitalShanghai201318China
| | - Wenyan Kang
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Dan Li
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghai200030China
- Zhangjiang Institute for Advanced StudyShanghai Jiao Tong UniversityShanghai201203China
| | - Bo Sun
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai201210China
- State Key Laboratory of Chemical BiologyShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai200032China
- Shanghai Academy of Natural Sciences (SANS)Fudan UniversityShanghai200433China
| |
Collapse
|
36
|
López-García P, Tejero-Ojeda MM, Vaquero ME, Carrión-Vázquez M. Current amyloid inhibitors: Therapeutic applications and nanomaterial-based innovations. Prog Neurobiol 2025; 247:102734. [PMID: 40024279 DOI: 10.1016/j.pneurobio.2025.102734] [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: 09/03/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Amyloid proteins have long been in the spotlight for being involved in many degenerative diseases including Alzheimer´s, Parkinson´s or type 2 diabetes, which currently cannot be prevented and for which there is no effective treatment or cure. Here we provide a comprehensive review of inhibitors that act directly on the amyloidogenic pathway (at the monomer, oligomer or fibril level) of key pathological amyloids, focusing on the most representative amyloid-related diseases. We discuss the latest advances in preclinical and clinical trials, focusing on cutting-edge developments, particularly on nanomaterials-based inhibitors, which offer unprecedented opportunities to address the complexity of protein misfolding disorders and are revolutionizing the landscape of anti-amyloid therapeutics. Notably, nanomaterials are impacting critical areas such as bioavailability, penetrability and functionality of compounds currently used in biomedicine, paving the way for more specific therapeutic solutions tailored to various amyloid-related diseases. Finally, we highlight the window of opportunity opened by comparative analysis with so-called functional amyloids for the development of innovative therapeutic approaches for these devastating diseases.
Collapse
|
37
|
Luo Y, Xiang Y, Liu J, Hu Y, Guo J. A Multi-omics Framework Based on Machine Learning as a Predictor of Cognitive Impairment Progression in Early Parkinson's Disease. Neurol Ther 2025; 14:643-658. [PMID: 39985630 PMCID: PMC11906927 DOI: 10.1007/s40120-025-00716-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
INTRODUCTION Cognitive impairment (CI) is a common non-motor symptom of Parkinson's disease (PD). However, the diagnosis and prediction of CI progression in PD remain challenging. We aimed to explore a multi-omics framework based on machine learning integrating comprehensive radiomics, cerebrospinal fluid biomarkers, and genetics information to identify CI progression in early PD. METHODS Patients were first diagnosed with PD without CI at baseline. According to whether CI progressed within 5 years, patients were divided into two groups: PD without CI and PD with CI. Radiomics signatures were extracted from patients' T1-weighted MRI. We used machine learning methods to construct radiomics, hybrid, and multi-omics models in the training set and validated the models in the testing set. RESULT In the two groups, we found 7, 23, and 25 radiomics signatures with significant differences in the parietal, temporal, and frontal lobes, respectively. The radiomics model using the 25 signatures of the frontal lobe had an accuracy of 0.833 and an AUC (area under the curve) of 0.879 to predict CI progression. In addition, the hybrid model fused with the cerebrospinal fluid Aβ level had an accuracy of 0.867 and an AUC of 0.916. In our study, the multi-omics model showed the best predictive performance. The accuracy of the multi-omics model was 0.900, and the average AUC value after five-fold cross-validation was 0.928. CONCLUSION Radiomics signatures have a recognition effect in the CI progression in early PD. Multi-omics frameworks combining radiomics, cerebrospinal fluid biomarkers, and genetic information may be a potential predictor of CI progression in PD.
Collapse
Affiliation(s)
- Yang Luo
- Department of Neurology, XiangYa Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - YaQin Xiang
- Department of Neurology, XiangYa Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - JiaBin Liu
- Department of Neurology, XiangYa Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - YuXuan Hu
- Department of Neurology, XiangYa Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - JiFeng Guo
- Department of Neurology, XiangYa Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China.
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
38
|
Samidurai M, Chennakesavan K, Sarkar S, Malovic E, Nguyen HM, Singh L, Kumar A, Ealy A, Janarthanam C, Palanisamy BN, Kondru N, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Zhang H, Wulff H, Kanthasamy A. KCa3.1 Contributes to Neuroinflammation and Nigral Dopaminergic Neurodegeneration in Experimental models of Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643982. [PMID: 40166152 PMCID: PMC11956954 DOI: 10.1101/2025.03.18.643982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic neuroinflammation and misfolded α-synuclein (αSyn) have been identified as key pathological correlates driving Parkinson's disease (PD) pathogenesis; however, the contribution of ion channels to microglia activation in the context of α-synucleinopathy remains elusive. Herein, we show that KCa3.1, a calcium-activated potassium channel, is robustly upregulated within microglia in multiple preclinical models of PD and, most importantly, in human PD and dementia with Lewy bodies (DLB) brains. Pharmacological inhibition of KCa3.1 via senicapoc or TRAM-34 inhibits KCa3.1 channel activity and the associated reactive microglial phenotype in response to aggregated αSyn, as well as ameliorates of PD like pathology in diverse PD mouse models. Additionally, proteomic and transcriptomic profiling of microglia revealed that senicapoc ameliorates aggregated αSyn-induced, inflammation-associated pathways and dysregulated metabolism in primary microglial cells. Mechanistically, FYN kinase in a STAT1 dependent manner regulates KCa3.1 mediated the microglial reactive activation phenotype after α-synucleinopathy. Moreover, reduced neuroinflammation and subsequent PD-like neuropathology were observed in SYN AAV inoculated KCa3.1 knockout mice. Together, these findings suggest that KCa3.1 inhibition represents a novel therapeutic strategy for treating patients with PD and related α-synucleinopathies.
Collapse
|
39
|
Li Y, Torok J, Ding J, Wang N, Lau C, Kulkarni S, Anand C, Tran J, Cheng M, Lo C, Lu B, Sun Y, Yang X, Raj A, Peng C. Distinguish risk genes functioning at presynaptic or postsynaptic regions and key connectomes associated with pathological α-synuclein spreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642462. [PMID: 40161679 PMCID: PMC11952395 DOI: 10.1101/2025.03.11.642462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Previous studies have suggested that pathological α-synuclein (α-Syn) mainly transmits along the neuronal network, but several key questions remain unanswered: (1) How many and which connections in the connectome are necessary for predicting the progression of pathological α-Syn? (2) How to identify risk gene that affects pathology spreading functioning at presynaptic or postsynaptic regions, and are these genes enriched in different cell types? Here, we addressed these key questions with novel mathematical models. Strikingly, the spreading of pathological α-Syn is predominantly determined by the key subnetworks composed of only 2% of the strongest connections in the connectome. We further explored the genes that are responsible for the selective vulnerability of different brain regions to transmission to distinguish the genes that play roles in presynaptic from those in postsynaptic regions. Those risk genes were significantly enriched in microglial cells of presynaptic regions and neurons of postsynaptic regions. Gene regulatory network analyses were then conducted to identify 'key drivers' of genes responsible for selective vulnerability and overlapping with Parkinson's disease risk genes. By identifying and discriminating between key gene mediators of transmission operating at presynaptic and postsynaptic regions, our study has demonstrated for the first time that these are functionally distinct processes.
Collapse
|
40
|
Biggs KE, Fikse EN, Anderson FL, Kettenbach AN, Havrda MC. Coronin1A Regulates the Trafficking of Alpha Synuclein in Microglia. J Neurosci 2025; 45:e1337242025. [PMID: 39837661 PMCID: PMC11905355 DOI: 10.1523/jneurosci.1337-24.2025] [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: 07/08/2024] [Revised: 12/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025] Open
Abstract
Microglia respond to cytotoxic protein aggregates associated with the progression of neurodegenerative disease. Pathological protein aggregates activate the microglial NLRP3 inflammasome resulting in proinflammatory signaling, secretion, and potentially pyroptotic cell death. We characterized mixed sex primary mouse microglia exposed to microbial stressors and alpha synuclein preformed fibrils (αsyn PFFs) to identify cellular mechanisms related to Parkinson's disease. Microglia package and release the endosome fate regulator Coronin1A (Coro1A) in EVs in an Nlrp3-dependent manner in widely used experimental activation conditions. We were surprised to find that Coro1A packaging and release was not Nlrp3-dependent in αsyn PFF exposure conditions. Coro1A-/- microglia exposed to αsyn PFFs trafficked more αsyn to the lysosomal compartment increasing lysosomal membrane permeabilization. This corresponds to a decrease in αsyn released in EVs suggesting that Coro1A functions to shunt pathological proteins to a secretory pathway to attenuate lysosomal stress. αsyn PFF-driven lysosomal stress resulting from Coro1a loss was associated with enhanced cytotoxicity. Intrinsic apoptosis signaling was unaffected, but we observed elevated cytosolic cathepsin B and the presence of a cathepsin-associated 55 kD PARP cleavage product. Postmortem analysis of the PD mesencephalon supported a role for Coro1A in microglia, revealing elevated levels of Coro1A protein in human PD brains compared with those of healthy donors. Findings are relevant to the distribution of pathological αsyn and indicate that Coro1a protects microglia from lysosomal overload, inflammasome activation, and pyroptotic demise.
Collapse
Affiliation(s)
- Karl E Biggs
- Departments of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
- Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
| | - Emma N Fikse
- Departments of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
| | - Faith L Anderson
- Departments of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
| | - Arminja N Kettenbach
- Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03766
| | - Matthew C Havrda
- Departments of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03766
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03766
| |
Collapse
|
41
|
Lin Q, Cao D, Li W, Zhang Y, Li Y, Liu P, Huang X, Huang K, Gong Q, Zhou D, An D. Connectome architecture for gray matter atrophy and surgical outcomes in temporal lobe epilepsy. Epilepsia 2025. [PMID: 40056026 DOI: 10.1111/epi.18343] [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: 11/03/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Temporal lobe epilepsy (TLE) has been recognized as a network disorder with widespread gray matter atrophy. However, the role of connectome architecture in shaping morphological alterations and identifying atrophy epicenters remains unclear. Furthermore, individualized modeling of atrophy epicenters and their potential clinical applications have not been well established. This study aims to explore how gray matter atrophy correlates with normal connectome architecture, identify potential atrophy epicenters, and employ individualized modeling approach to evaluate the impact of different epicenter patterns on surgical outcomes in patients with TLE. METHODS This study utilized anatomic MRI data from 126 refractory TLE patients who underwent anterior temporal lobectomy and 60 healthy controls (HCs), along with normative functional and structural connectome data, to investigate the relationship between gray matter volume (GMV) changes and functional or structural connectivity. Two models were employed to identify atrophy epicenters: a data-driven approach evaluating nodal and neighbor atrophy rankings, and a network diffusion model (NDM) simulating the spread of pathology from different seed regions. K-means clustering was applied in patient-tailored modeling to uncover distinct epicenter subtypes. RESULTS Our findings indicate that the pattern of gray matter atrophy in TLE is constrained primarily by structural connectivity rather than by functional connectivity. Using the structural connectome, we pinpointed the hippocampus and adjacent temporo-limbic regions as key atrophy epicenters. The patient-tailored modeling revealed significant variability in epicenter distribution, allowing us to categorize them into two distinct subtypes. Notably, patients in subtype 2, with epicenters localized to the ipsilateral temporal pole and medial temporal lobe, exhibited significantly higher seizure-free rates compared to patients in subtype 1, whose epicenters situated in frontocentral regions. SIGNIFICANCE These findings highlight the central role of structural connectivity in shaping TLE-related morphological changes. Individualized epicenter modeling may enhance surgical decisions and improve prognostic stratification in TLE management.
Collapse
Affiliation(s)
- Qiuxing Lin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyang Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingying Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuming Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiwen Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiang Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kailing Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiyong Gong
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongmei An
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Sirerol-Piquer MS, Perez-Villalba A, Duart-Abadia P, Belenguer G, Gómez-Pinedo U, Blasco-Chamarro L, Carrillo-Barberà P, Pérez-Cañamás A, Navarro-Garrido V, Dehay B, Vila M, Vitorica J, Pérez-Sánchez F, Fariñas I. Age-dependent progression from clearance to vulnerability in the early response of periventricular microglia to α-synuclein toxic species. Mol Neurodegener 2025; 20:26. [PMID: 40038767 PMCID: PMC11881471 DOI: 10.1186/s13024-025-00816-1] [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: 05/18/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
Cytoplasmic alpha-synuclein (αSyn) aggregates are a typical feature of Parkinson's disease (PD). Extracellular insoluble αSyn can induce pathology in healthy neurons suggesting that PD neurodegeneration may spread through cell-to-cell transfer of αSyn proteopathic seeds. Early pro-homeostatic reaction of microglia to toxic forms of αSyn remains elusive, which is especially relevant considering the recently uncovered microglial molecular diversity. Here, we show that periventricular microglia of the subependymal neurogenic niche monitor the cerebrospinal fluid and can rapidly phagocytize and degrade different aggregated forms of αSyn delivered into the lateral ventricle. However, this clearing ability worsens with age, leading to an increase in microglia with aggregates in aged treated mice, an accumulation also observed in human PD samples. We also show that exposure of aged microglia to aggregated αSyn isolated from human PD samples results in the phosphorylation of the endogenous protein and the generation of αSyn seeds that can transmit the pathology to healthy neurons. Our data indicate that while microglial phagocytosis rapidly clears toxic αSyn, aged microglia can contribute to synucleinopathy spreading.
Collapse
Affiliation(s)
- Mª Salomé Sirerol-Piquer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain.
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain.
| | - Ana Perez-Villalba
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
- L.A.B.P. (Laboratory of Animal Behavior Phenotype), Facultad de Psicología. UCV, Valencia, Spain
| | - Pere Duart-Abadia
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Germán Belenguer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Institute of Neurosciences, Hospital Clínico San Carlos Health Research Institute, Universidad Complutense de Madrid, Madrid, Spain
| | - Laura Blasco-Chamarro
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Pau Carrillo-Barberà
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Azucena Pérez-Cañamás
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Victoria Navarro-Garrido
- Instituto de Biomedicina de Sevilla (IBiS), Universidad de Sevilla, Seville, Spain
- Departamento Bioquímica y Biología Molecular, Universidad de Sevilla, Seville, Spain
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F-33000, France
| | - Miquel Vila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d´Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Javier Vitorica
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Universidad de Sevilla, Seville, Spain
- Departamento Bioquímica y Biología Molecular, Universidad de Sevilla, Seville, Spain
| | - Francisco Pérez-Sánchez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain.
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Burjassot, Spain.
| |
Collapse
|
43
|
Nuermaimaiti M, Ishikawa KI, Oyama G, Nonaka R, Shiga T, Jo T, Tsunemi T, Nakamura R, Krüger R, Akamatsu W, Hattori N. Human induced pluripotent stem cell-derived dopaminergic neurons release alpha-synuclein through neuronal activity. Neurosci Res 2025; 212:105-114. [PMID: 39617169 DOI: 10.1016/j.neures.2024.11.007] [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: 07/23/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024]
Abstract
Lewy body diseases, including Parkinson's disease (PD), are characterized by the spread of alpha-synuclein (αSyn) between neurons across synapses, a process crucial for understanding their pathophysiology and developing effective treatments. In this study, we aimed to investigate the role of neuronal activity in releasing αSyn from human induced pluripotent stem cell-derived dopaminergic neurons. We examined human induced pluripotent stem cell-derived dopaminergic neurons, both healthy and those with the αSyn gene mutation associated with PD. We employed pharmacological agents and optogenetic techniques and demonstrated that increased neuronal activity, induced by bicuculline or optogenetic stimulation, significantly enhances αSyn release. However, suppression of neuronal activity with cyanquixaline reduces αSyn secretion. These findings underscore the pivotal role of neuronal activity in αSyn transmission between neurons, showing its potential impact on the spread of Lewy pathology in patients with neurodegenerative diseases like PD. Therefore, this study advances our understanding of PD and opens new avenues for therapeutic strategies to mitigate Lewy body disease progression.
Collapse
Affiliation(s)
- Maierdanjiang Nuermaimaiti
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Clinical Data of Parkinson's Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kei-Ichi Ishikawa
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan.
| | - Genko Oyama
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Risa Nonaka
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Laboratory of Molecular Biology, Faculty of Pharmacy, Juntendo University, Chiba, Japan
| | - Takahiro Shiga
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takayuki Jo
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Taiji Tsunemi
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Ryota Nakamura
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Clinical Data of Parkinson's Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
44
|
Yu L, Li X, Shi T, Li N, Zhang D, Liu X, Xiao Y, Liu X, Petersen RB, Xue W, Yu YV, Hu DS, Xu L, Chen H, Zheng L, Huang K, Peng A. Identification of novel phenolic inhibitors from traditional Chinese medicine against toxic α-synuclein aggregation via regulating phase separation. Int J Biol Macromol 2025; 297:139875. [PMID: 39818366 DOI: 10.1016/j.ijbiomac.2025.139875] [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: 11/27/2024] [Revised: 12/30/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder without cure, is characterized by the pathological aggregation of α-synuclein (α-Syn) in Lewy bodies. Classic deposition pathway and condensation pathway contribute to α-Syn aggregation, and liquid-liquid phase separation is the driving force for condensate formation, which subsequently undergo liquid-solid phase separation to form toxic fibrils. Traditional Chinese Medicine (TCM) has a long history in treating neurodegenerative disease; herein, we identified chemicals from herbs that inhibit α-Syn aggregation. We screened commonly prescribed TCMs for PD from the CNKI database and registered patents, 13 chemicals were identified in the TCMSP databases as candidate inhibitors, among which three phenols, forsythoside B (FTSB), echinacoside (ECH), and 4-hydroxyindole (C4-OH) efficiently inhibit α-Syn aggregation. Moreover, FTSB and ECH increase α-Syn fluidity within condensates, inhibit α-Syn transition into amyloid fibrils and reduce fibril-induced toxicity in SH-SY5Y cells. Importantly, they disaggregated preformed α-Syn amyloid fibrils. Notably, in an α-Syn overexpressing NL5901 C. elegans PD model, either FTSB or ECH treatment significantly extended the lifespan and improved the PD-like movement disorders, both in the preventive and therapeutic treatment approaches, by reducing toxic α-Syn inclusion formation and improving the fluidity of α-Syn. Together, we offer new therapeutic candidates targeting phase separation-associated aggregation for PD.
Collapse
Affiliation(s)
- Linwei Yu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Li
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyi Shi
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ning Li
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Donge Zhang
- Wuhan Third hospital, Tongren Hospital of Wuhan University, 241 Pengliuyang Road, Wuhan 430060, China
| | - Xikai Liu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yushuo Xiao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinran Liu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI 48859, USA
| | - Weikang Xue
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430070, China
| | - Yanxun V Yu
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430070, China
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Li Xu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Tongji-Rong Cheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Anlin Peng
- Wuhan Third hospital, Tongren Hospital of Wuhan University, 241 Pengliuyang Road, Wuhan 430060, China.
| |
Collapse
|
45
|
Ma Y, Erb ML, Moore DJ. Aging, cellular senescence and Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2025; 15:239-254. [PMID: 39973488 DOI: 10.1177/1877718x251316552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, affecting 1-2% of people over age 65. The risk of developing PD dramatically increases with advanced age, indicating that aging is likely a driving factor in PD neuropathogenesis. Several age-associated biological changes are also hallmarks of PD neuropathology, including mitochondrial dysfunction, oxidative stress, and neuroinflammation. Accumulation of senescent cells is an important feature of aging that contributes to age-related diseases. How age-related cellular senescence affects brain health and whether this phenomenon contributes to neuropathogenesis in PD is not yet fully understood. In this review, we highlight hallmarks of aging, including mitochondrial dysfunction, loss of proteostasis, genomic instability and telomere attrition in relation to well established PD neuropathological pathways. We then discuss the hallmarks of cellular senescence in the context of neuroscience and review studies that directly examine cellular senescence in PD. Studying senescence in PD presents challenges and holds promise for advancing our understanding of disease mechanisms, which could contribute to the development of effective disease-modifying therapeutics. Targeting senescent cells or modulating the senescence-associated secretory phenotype (SASP) in PD requires a comprehensive understanding of the complex relationship between PD pathogenesis and cellular senescence.
Collapse
Affiliation(s)
- Yue Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Madalynn L Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
46
|
Fielding L, Menard MA, Roth J, Iuliano M, Dehay B, Aguilar-Calvo P, Volpicelli-Daley LA. Current safety recommendations for handling mouse and human αsynuclein pre-formed fibrils. Neurobiol Dis 2025; 206:106820. [PMID: 39889858 DOI: 10.1016/j.nbd.2025.106820] [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: 11/25/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
α-Synuclein (α-syn) can form amyloid fibrils. Lewy bodies and Lewy neurites containing aggregated α-syn are pathological markers of Parkinson's Disease and Dementia with Lewy Bodies. To better understand the role of pathological α-syn in disease, many labs use α-syn preformed fibrils (PFFs). Neurons take up the PFFs, which act as seeds to corrupt endogenously expressed α-syn, inducing it to form aggregates very similar to those found in diseased brains. The PFFs are typically generated using recombinant mouse or human α-syn. α-Syn fibrils can also be extracted or amplified from brain tissue extracts, cerebrospinal fluid, or skin biopsies from patients with known synucleinopathy. The PFFs are then added to cell culture, or injected into rodents or primates to induce pathology. Because PFFs can corrupt endogenous α-syn, researchers should adhere to strict safety protocols when handling PFFs to minimize potential exposures. Our group consulted with biosafety professionals at the University of Alabama at Birmingham (UAB) to identify potential risks related to working with α-syn PFFs and offer containment controls to mitigate those risks. Potential exposures include pipetting, opening tubes, and sonication of the PFFs to generate fragments, all of which could potentially generate aerosols. Here, we outline best practices for the safe conduct of research with α-syn fibrils, including personal protective equipment and decontamination procedures. We highlight steps in which extra precautions should be taken and how to minimize exposure and potential risk associated with use of PFFs in scientific research.
Collapse
Affiliation(s)
- Lauren Fielding
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, 1719 6th Avenue South, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Marissa A Menard
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, 1719 6th Avenue South, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Justin Roth
- Environmental Health and Safety, 933 19(th) Street South, University of Alabama at Birmingham, Birmingham, AL 35205, USA.
| | - Maria Iuliano
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511 and Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA; Aligning Science Across Parkinson's (ASA) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | | | - Patricia Aguilar-Calvo
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, 1719 6th Avenue South, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, 1719 6th Avenue South, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Aligning Science Across Parkinson's (ASA) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
47
|
Szegvari E, Holec SAM, Woerman AL. Limitations and Applications of Rodent Models in Tauopathy and Synucleinopathy Research. J Neurochem 2025; 169:e70021. [PMID: 40026260 PMCID: PMC11874209 DOI: 10.1111/jnc.70021] [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: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 03/05/2025]
Abstract
Rodent models that accurately recapitulate key aspects of human disease have long been fundamental to the successful development of clinical interventions. This is greatly underscored in the neurodegenerative disease field, where preclinical testing of anti-prion therapeutics against rodent-adapted prions resulted in the development of small molecules effective against rodent-adapted prions but not against human prions. These findings provided critical lessons for ongoing efforts to develop treatments for patients with neurodegenerative diseases caused by misfolding and accumulation of the proteins tau and α-synuclein, or tauopathies and synucleinopathies, respectively. To avoid the potential pitfalls previously identified in the prion field, this review focuses on rodent models currently available to study tau and α-synuclein disease pathogenesis, emphasizing the strengths and limitations of each with the particular goal of better supporting preclinical research.
Collapse
Affiliation(s)
- Emma Szegvari
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| | - Sara A. M. Holec
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| | - Amanda L. Woerman
- Department of Microbiology, Immunology, & Pathology and Prion Research CenterColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
48
|
Almasi F, Abbasloo F, Soltani N, Dehbozorgi M, Moghadam Fard A, Kiani A, Ghasemzadeh N, Mesgari H, Zadeh Hosseingholi E, Payandeh Z, Rahmanpour P. Biology, Pathology, and Targeted Therapy of Exosomal Cargoes in Parkinson's Disease: Advances and Challenges. Mol Neurobiol 2025:10.1007/s12035-025-04788-7. [PMID: 39998798 DOI: 10.1007/s12035-025-04788-7] [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: 09/21/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Parkinson's disease (PD) involves the loss of dopamine neurons and accumulation of alpha-synuclein (α-syn), leading to Lewy bodies. While α-syn-targeting immunotherapies show promise, clinical application is challenging. Emerging strategies include nano-platforms for targeted delivery and imaging, and cell-based therapies with patient-specific dopaminergic neurons, aiming to enhance treatment effectiveness despite challenges. Exosome-based methodologies are emerging as a promising area of research in PD due to their role in the spread of α-syn pathology. Exosomes are small extracellular vesicles that can carry misfolded α-syn and transfer it between cells, contributing to the progression of PD. They can be isolated from biological fluids such as blood and cerebrospinal fluid, making them valuable biomarkers for the disease. Additionally, engineering exosomes to deliver therapeutic agents, including small molecules, RNA, or proteins, offers a novel approach for targeted therapy, capitalizing on their natural ability to cross the blood-brain barrier (BBB). Ongoing studies are evaluating the safety and efficacy of these engineered exosomes in clinical settings. This review explores the role of exosomes in PD, focusing on their potential for diagnosis, treatment, and understanding of pathology. It highlights advancements and future directions in using exosomes as biomarkers and therapeutic tools.
Collapse
Affiliation(s)
- Faezeh Almasi
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, 45139-56184, Iran.
| | - Faeze Abbasloo
- Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Soltani
- Center for Gene Regulation in Health and Disease, Department of Biological Sciences, Cleveland State University, Cleveland, OH, 44115, USA
| | - Masoud Dehbozorgi
- Faculty of Medicine, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH Aachen), Aachen City, Germany
| | | | - Arash Kiani
- Yasuj University of Medical Sciences, Yasuj, Iran
| | - Nasim Ghasemzadeh
- School of Natural Sciences and Mathematics, University of Dallas, Richardson, TX, USA
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Elaheh Zadeh Hosseingholi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran.
| | - Zahra Payandeh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41346, Gothenburg, Sweden.
| | | |
Collapse
|
49
|
Tullo S, Park J, Gallino D, Park M, Mar K, Novikov V, Sandoval Contreras R, Patel R, Del Cid-Pellitero E, Fon EA, Luo W, Shlaifer I, Durcan TM, Prado MAM, Prado VF, Devenyi GA, Chakravarty MM. Female mice exhibit resistance to disease progression despite early pathology in a transgenic mouse model inoculated with alpha-synuclein fibrils. Commun Biol 2025; 8:288. [PMID: 39987244 PMCID: PMC11846974 DOI: 10.1038/s42003-025-07680-1] [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: 06/07/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025] Open
Abstract
Despite known sex differences in human synucleinopathies such as Parkinson's disease, the impact of sex on alpha-synuclein pathology in mouse models has been largely overlooked. To address this need, we examine sex differences in whole brain signatures of neurodegeneration due to aSyn toxicity in the M83 mouse model using longitudinal magnetic resonance imaging (MRI; T1-weighted; 100 μm3 isotropic voxel; -7, 30, 90 and 120 days post-injection [dpi]; n ≥ 8 mice/group/sex/time point). To initiate aSyn spreading, M83 mice are inoculated with recombinant human aSyn preformed fibrils (Hu-PFF) or phosphate buffered saline in the right striatum. We observe more aggressive neurodegenerative profiles over time for male Hu-PFF-injected mice when examining voxel-wise trajectories. However, at 90 dpi, we observe widespread patterns of neurodegeneration in the female Hu-PFF-injected mice. These differences are not accompanied by any differences in motor symptom onset between the sexes. However, male Hu-PFF-injected mice reached their humane endpoint sooner. These findings suggest that post-motor symptom onset, despite accelerated disease trajectories for male Hu-PFF-injected mice, neurodegeneration may appear sooner in the female Hu-PFF-injected mice (prior to motor symptomatology). These findings suggest that sex-specific synucleinopathy phenotypes urgently need to be considered to improve our understanding of neuroprotective and neurodegenerative mechanisms.
Collapse
Affiliation(s)
- Stephanie Tullo
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada.
| | - Janice Park
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
| | - Daniel Gallino
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
| | - Megan Park
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
| | - Kristie Mar
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
| | - Vladislav Novikov
- Robarts Research Institute, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
| | - Rodrigo Sandoval Contreras
- Robarts Research Institute, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
| | - Raihaan Patel
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
- Department of Biological & Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Esther Del Cid-Pellitero
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Wen Luo
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine, The University of Western Ontario, London, ON, Canada
| | - Gabriel A Devenyi
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - M Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Cerebral Imaging Center, Douglas Research Center, McGill University, Verdun, QC, Canada.
- Department of Biological & Biomedical Engineering, McGill University, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
50
|
Taguchi K, Watanabe Y, Tanaka M. SENP2-based N-terminal truncation of α-synuclein in Lewy pathology propagation. iScience 2025; 28:111935. [PMID: 40028275 PMCID: PMC11869972 DOI: 10.1016/j.isci.2025.111935] [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: 09/30/2024] [Revised: 11/10/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
α-Synuclein (αSyn) is a major component of Lewy bodies (LBs) and Lewy neurites (LNs), which are pathological features of Parkinson's disease (PD) and dementia with Lewy bodies. In the PD brain, with disease progression, LB/LN formation is propagated from the lower brainstem to the cerebral cortex. Prion-like cell-to-cell seed transmission has been implicated as an underlying mechanism for Lewy-pathology propagation. However, the biochemical properties and production mechanism of those pathogenic seeds are unelucidated. In this study, we ascertained that the seeds released from pathological neurons that harbor LB/LN-like aggregates have the N-terminally truncated form of αSyn. This N-terminal truncation is directly catalyzed by SENP2, which is a well-known deSUMOylation enzyme. After SENP2 processing of recombinant αSyn, the SDS-resistant high-molecular oligomer formation was promoted in vitro. Inhibition of SENP2 activity suppressed aggregate formation and propagation in cultured neurons and mouse brains. Thus, SENP2 might be a therapeutic target in LB diseases.
Collapse
Affiliation(s)
- Katsutoshi Taguchi
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Yoshihisa Watanabe
- Department of Basic Geriatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Masaki Tanaka
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|