1
|
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
|
2
|
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
|
3
|
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
|
4
|
Theologidis V, Ferreira SA, Jensen NM, Gomes Moreira D, Ahlgreen OA, Hansen MW, Rosenberg ED, Richner M, Faress I, Gram H, Jensen PH, Borghammer P, Nyengaard JR, Romero-Ramos M, Vægter CB, van de Berg WDJ, Van Den Berge N, Jan A. Bradykinesia and postural instability in a model of prodromal synucleinopathy with α-synuclein aggregation initiated in the gigantocellular nuclei. Acta Neuropathol Commun 2025; 13:32. [PMID: 39962601 PMCID: PMC11834571 DOI: 10.1186/s40478-025-01948-7] [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: 10/04/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
α-Synuclein (aSyn) accumulation within the extra-nigral neuronal populations in the brainstem, including the gigantocellular nuclei (GRN/Gi) of reticular formation, is a recognized feature during the prodromal phase of Parkinson disease (PD). Accordingly, there is a burgeoning interest in animal model development for understanding the pathological significance of extra-nigral synucleinopathy, in relation to motor and/or non-motor symptomatology in PD. Here, we report an experimental paradigm for the induction of aSyn aggregation in brainstem, with stereotaxic delivery of pre-formed fibrillar (PFF) aSyn in the pontine GRN of transgenic mice expressing the mutant human Ala53Thr aSyn (M83 line). Our data show that PFF aSyn-induced aggregate pathology in GRN and distinct nuclei of subcortical motor system leads to progressive decline in home cage activity, which was accompanied by postural instability and impaired motor coordination. The progressive accumulation of aSyn pathology in brainstem and motor neurons in lumbar spinal cord heralded the onset of a moribund stage, which culminated in impaired survival. Collectively, our observations suggest an experimental framework for studying the pathological significance of aSyn aggregation in GRN in relation to features of movement disability in PD. With further refinements, we anticipate that this model holds promise as a test-bed for translational research in PD and related disorders.
Collapse
Affiliation(s)
- Vasileios Theologidis
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
- Core Center for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
| | - Sara A Ferreira
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Nanna M Jensen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Diana Gomes Moreira
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
| | - Ole A Ahlgreen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Mads W Hansen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Emilie D Rosenberg
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Mette Richner
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Islam Faress
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Hjalte Gram
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Per Borghammer
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
| | - Jens R Nyengaard
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
- Core Center for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Christian B Vægter
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Nathalie Van Den Berge
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
- Core Center for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 35, 8200, Aarhus, Denmark
| | - Asad Jan
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus, Denmark.
| |
Collapse
|
5
|
Mesentier-Louro LA, Goldman C, Ndayisaba A, Buonfiglioli A, Rooklin RB, Schuldt BR, Uchitelev A, Khurana V, Blanchard JW. Cholesterol-mediated Lysosomal Dysfunction in APOE4 Astrocytes Promotes α-Synuclein Pathology in Human Brain Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637107. [PMID: 39975381 PMCID: PMC11839026 DOI: 10.1101/2025.02.09.637107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The pathological hallmark of neurodegenerative disease is the aberrant post-translational modification and aggregation of proteins leading to the formation of insoluble protein inclusions. Genetic factors like APOE4 are known to increase the prevalence and severity of tau, amyloid, and α-Synuclein inclusions. However, the human brain is largely inaccessible during this process, limiting our mechanistic understanding. Here, we developed an iPSC-based 3D model that integrates neurons, glia, myelin, and cerebrovascular cells into a functional human brain tissue (miBrain). Like the human brain, we found pathogenic phosphorylation and aggregation of α-Synuclein is increased in the APOE4 miBrain. Combinatorial experiments revealed that lipid-droplet formation in APOE4 astrocytes impairs the degradation of α-synuclein and leads to a pathogenic transformation that seeds neuronal inclusions of α-Synuclein. Collectively, this study establishes a robust model for investigating protein inclusions in human brain tissue and highlights the role of astrocytes and cholesterol in APOE4-mediated pathologies, opening therapeutic opportunities.
Collapse
Affiliation(s)
- Louise A. Mesentier-Louro
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Camille Goldman
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Alain Ndayisaba
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alice Buonfiglioli
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Rikki B. Rooklin
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Braxton R. Schuldt
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Abigail Uchitelev
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Macaulay Honors College at Hunter College, New York, NY, USA
| | - Vikram Khurana
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Joel W. Blanchard
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Lead contact
| |
Collapse
|
6
|
Farzadfard A, Mason TO, Kunka A, Mohammad‐Beigi H, Bjerregaard‐Andersen K, Folke J, Aznar S, Kallunki P, Buell AK. The Amplification of Alpha-Synuclein Amyloid Fibrils is Suppressed under Fully Quiescent Conditions. Angew Chem Int Ed Engl 2025; 64:e202419173. [PMID: 39648869 PMCID: PMC11811685 DOI: 10.1002/anie.202419173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Seed amplification assays (SAAs) are a promising avenue for the early diagnosis of neurodegenerative diseases. However, when amplifying fibrils from patient-derived samples in multiwell plates, it is currently highly challenging to accurately quantify the aggregates. It is therefore desirable to transfer such assays into a digital format in microemulsion droplets to enable direct quantification of aggregate numbers. To achieve transfer from conventional plate-based to the microfluidic digital format, effective seed amplification needs to be achieved inside the microdroplets. Therefore, we establish a new set of assay conditions that enable highly efficient seed amplification in plates without any shaking. However, the same set of conditions displayed a very different behavior upon transfer to a microfluidic platform where no amplification was observed. We demonstrate that this is caused by the suppression of all secondary processes that could amplify the seeds in the complete absence of mechanical perturbations inside the microdroplets. We further show that the amplification inside droplets can be achieved by subjecting the microemulsions to high-frequency vibrations using a piezo device. Taken together, our results provide novel insights into the physical requirements of alpha-synuclein seed amplification and demonstrate a pathway towards the development of effective digital SAAs.
Collapse
Affiliation(s)
- Azad Farzadfard
- Protein Biophysics groupDepartment of Biotechnology and BiomedicineTechnical University of DenmarkSøltofts Plads, Building 2272800, Kgs.Lyngby
- H. Lundbeck A/SCarl Jacobsens Vej 222500KøbenhavnDenmark
| | - Thomas O. Mason
- Protein Biophysics groupDepartment of Biotechnology and BiomedicineTechnical University of DenmarkSøltofts Plads, Building 2272800, Kgs.Lyngby
| | - Antonin Kunka
- Protein Biophysics groupDepartment of Biotechnology and BiomedicineTechnical University of DenmarkSøltofts Plads, Building 2272800, Kgs.Lyngby
| | - Hossein Mohammad‐Beigi
- Protein Biophysics groupDepartment of Biotechnology and BiomedicineTechnical University of DenmarkSøltofts Plads, Building 2272800, Kgs.Lyngby
| | | | - Jonas Folke
- Centre for Neuroscience and StereologyDepartment of NeurologyBispebjerg-Frederiksberg HospitalUniversity Hospital of CopenhagenNielsine Nielsen Vej 6B, Entrance 11BDK-2400Copenhagen NV
| | - Susana Aznar
- Centre for Neuroscience and StereologyDepartment of NeurologyBispebjerg-Frederiksberg HospitalUniversity Hospital of CopenhagenNielsine Nielsen Vej 6B, Entrance 11BDK-2400Copenhagen NV
| | - Pekka Kallunki
- H. Lundbeck A/SCarl Jacobsens Vej 222500KøbenhavnDenmark
| | - Alexander K. Buell
- Protein Biophysics groupDepartment of Biotechnology and BiomedicineTechnical University of DenmarkSøltofts Plads, Building 2272800, Kgs.Lyngby
| |
Collapse
|
7
|
Haikal C, Winston GM, Kaplitt MG. Cognitive dysfunction in animal models of human lewy-body dementia. Front Aging Neurosci 2024; 16:1369733. [PMID: 39104707 PMCID: PMC11298446 DOI: 10.3389/fnagi.2024.1369733] [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: 01/12/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Cognitive impairments are a common feature of synucleinopathies such as Parkinson's Disease Dementia and Dementia with Lewy Bodies. These pathologies are characterized by accumulation of Lewy bodies and Lewy neurites as well as neuronal cell death. Alpha-synuclein is the main proteinaceous component of Lewy bodies and Lewy neurites. To model these pathologies in vivo, toxins that selectively target certain neuronal populations or different means of inducing alpha-synuclein aggregation can be used. Alpha-synuclein accumulation can be induced by genetic manipulation, viral vector overexpression or the use of preformed fibrils of alpha-synuclein. In this review, we summarize the cognitive impairments associated with different models of synucleinopathies and relevance to observations in human diseases.
Collapse
Affiliation(s)
- Caroline Haikal
- Weill Cornell Medicine, Department of Neurological Surgery, New York, NY, United States
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, United States
| | - Graham M. Winston
- Weill Cornell Medicine, Department of Neurological Surgery, New York, NY, United States
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, United States
| | - Michael G. Kaplitt
- Weill Cornell Medicine, Department of Neurological Surgery, New York, NY, United States
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
8
|
Massey RS, Johri S, Chan D, Holahan MR, Prakash R. Comparison of Aptamer and Antibody Bioreceptors in the OEGFET Biosensor Platform for Detecting α-Synuclein, a Parkinson's Biomarker. IEEE SENSORS LETTERS 2024; 8:1-4. [DOI: 10.1109/lsens.2024.3413575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Roslyn S. Massey
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Srishti Johri
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Dennis Chan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Matthew R. Holahan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
9
|
Karim MR, Gasparini E, Tiegs E, Schlichte R, Vermilyea SC, Lee MK. Internalized α-synuclein fibrils become truncated and resist degradation in neurons while glial cells rapidly degrade α-synuclein fibrils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597615. [PMID: 38895363 PMCID: PMC11185753 DOI: 10.1101/2024.06.05.597615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Parkinson's disease (PD) and other α-synucleinopathies are characterized by the accumulation of α-synuclein (αS) pathology that can spread via the cell-to-cell transmission of αS aggregates. To better understand how various brain cells contribute to the spreading of αS pathology, we examined the metabolism of αS aggreges or pre-formed fibrils (PFFs) in neuronal and glial cells (microglia, astrocytes, and oligodendrocytes). In neurons, while the full-length αS rapidly disappeared following αS PFF uptake, truncated αS accumulated with a half-life of days rather than hours. Epitope mapping and fractionation studies indicate that αS PFF was truncated at the C-terminal region following uptake and remained insoluble/aggregated. In contrast, microglia and astrocytes rapidly metabolized αS PFF as the half-lives of αS PFF in these glial cells were <6 hours. Differential processing of αS by neurons was recapitulated in cell lines as differentiated CLU neuronal cell lines stably accumulate truncated αS while undifferentiated cells rapidly metabolize αS. Immunolocalization and subcellular fractionation studies show that internalized αS PFF is initially localized to endosomes followed by lysosomes. The lysosome is largely responsible for the degradation of internalized αS PFF as the inhibition of lysosomal function leads to the stabilization of αS in all cell types. Significantly, αS PFF causes lysosomal dysfunction in neurons. In summary, we show that neurons are inefficient in metabolizing internalized αS aggregates, partially because αS aggregates cause lysosomal dysfunction, potentially generating aggregation-prone truncated αS. In contrast, glial cells may protect neurons from αS aggregates by rapidly clearing αS aggregates.
Collapse
Affiliation(s)
- Md. Razaul Karim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Emilie Gasparini
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
| | - Elizabeth Tiegs
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Riley Schlichte
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Scott C. Vermilyea
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55414, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| |
Collapse
|
10
|
Laursen ALS, Olesen MV, Folke J, Brudek T, Knecht LH, Sotty F, Lambertsen KL, Fog K, Dalgaard LT, Aznar S. Systemic inflammation activates coagulation and immune cell infiltration pathways in brains with propagating α-synuclein fibril aggregates. Mol Cell Neurosci 2024; 129:103931. [PMID: 38508542 DOI: 10.1016/j.mcn.2024.103931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
Synucleinopathies are a group of diseases characterized by brain aggregates of α-synuclein (α-syn). The gradual accumulation of α-syn and the role of inflammation in early-stage pathogenesis remain poorly understood. We explored this interaction by inducing chronic inflammation in a common pre-clinical synucleinopathy mouse model. Three weeks post unilateral intra-striatal injections of human α-syn pre-formed fibrils (PFF), mice underwent repeated intraperitoneal injections of 1 mg/ml lipopolysaccharide (LPS) for 3 weeks. Histological examinations of the ipsilateral site showed phospho-α-syn regional spread and LPS-induced neutrophil recruitment to the brain vasculature. Biochemical assessment of the contralateral site confirmed spreading of α-syn aggregation to frontal cortex and a rise in intracerebral TNF-α, IL-1β, IL-10 and KC/GRO cytokines levels due to LPS. No LPS-induced exacerbation of α-syn pathology load was observed at this stage. Proteomic analysis was performed contralateral to the PFF injection site using LC-MS/MS. Subsequent downstream Reactome Gene-Set Analysis indicated that α-syn pathology alters mitochondrial metabolism and synaptic signaling. Chronic LPS-induced inflammation further lead to an overrepresentation of pathways related to fibrin clotting as well as integrin and B cell receptor signaling. Western blotting confirmed a PFF-induced increase in fibrinogen brain levels and a PFF + LPS increase in Iba1 levels, indicating activated microglia. Splenocyte profiling revealed changes in T and B cells, monocytes, and neutrophils populations due to LPS treatment in PFF injected animals. In summary, early α-syn pathology impacts energy homeostasis pathways, synaptic signaling and brain fibrinogen levels. Concurrent mild systemic inflammation may prime brain immune pathways in interaction with peripheral immunity.
Collapse
Affiliation(s)
- Anne-Line Strange Laursen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark; Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Mikkel Vestergaard Olesen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Jonas Folke
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Tomasz Brudek
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Luisa Harriet Knecht
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløwsvej 21-25, DK-5000, Odense, Denmark; Department of Neurology, Odense University Hospital, J.B. Winsløwsvej 4, Odense, Denmark; BRIDGE - Brain-Research-Inter-Disciplinary Guided Excellence, Department of Clinical Institute, University of Southern Denmark, Winsløwparken 19, Odense, Denmark.
| | - Karina Fog
- H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark.
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Susana Aznar
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| |
Collapse
|
11
|
Melachroinou K, Divolis G, Tsafaras G, Karampetsou M, Fortis S, Stratoulias Y, Papadopoulou G, Kriebardis AG, Samiotaki M, Vekrellis K. Endogenous Alpha-Synuclein is Essential for the Transfer of Pathology by Exosome-Enriched Extracellular Vesicles, Following Inoculation with Preformed Fibrils in vivo. Aging Dis 2024; 15:869-892. [PMID: 37548944 PMCID: PMC10917543 DOI: 10.14336/ad.2023.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/14/2023] [Indexed: 08/08/2023] Open
Abstract
The main pathological hallmark of Parkinson's disease (PD) and related synucleinopathies is the presence of intracellular proteinaceous aggregates, enriched in the presynaptic protein alpha-Synuclein (α-Syn). α-Syn association with exosomes has been previously documented both as a physiological process of secretion and as a pathological process of disease transmission, however, critical information about the mechanisms governing this interplay is still lacking. To address this, we utilized the α-Syn preformed fibril (PFF) mouse model of PD, as a source of brain-derived exosome-enriched extracellular vesicles (ExE-EVs) and assessed their pathogenic capacity following intrastriatal injections in host wild type (WT) mouse brain. We further investigated the impact of the fibrillar α-Syn on the exosomal cargo independent of the endogenous α-Syn, by isolating ExE-EVs from PFF-injected α-Syn knockout mice. Although PFF inoculation does not alter the morphology, size distribution, and quantity of brain-derived ExE-EVs, it triggers changes in the exosomal proteome related to synaptic and mitochondrial function, as well as metabolic processes. Importantly, we showed that the presence of the endogenous α-Syn is essential for the ExE-EVs to acquire a pathogenic capacity, allowing them to mediate disease transmission by inducing phosphorylated-α-Syn pathology. Notably, misfolded α-Syn containing ExE-EVs when injected in WT mice were able to induce astrogliosis and synaptic alterations in the host brain, at very early stages of α-Syn pathology, preceding the formation of the insoluble α-Syn accumulations. Collectively, our data suggest that exosomal cargo defines their ability to spread α-Syn pathology.
Collapse
Affiliation(s)
- Katerina Melachroinou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Georgios Divolis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - George Tsafaras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Mantia Karampetsou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Sotirios Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece.
| | - Yannis Stratoulias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Gina Papadopoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Kostas Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
12
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
13
|
Lee M, Barnes J, Vermilyea S, Meints J, Martinez H. Soluble and insoluble lysates from the human A53T mutant α-synuclein transgenic mouse model induces α-synucleinopathy independent of injection site. RESEARCH SQUARE 2024:rs.3.rs-3982325. [PMID: 38496623 PMCID: PMC10942550 DOI: 10.21203/rs.3.rs-3982325/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Pathological aggregation of a-synuclein (aS) is implicated in the pathogenesis of Parkinson's disease (PD) and other a-synucleinopathies. The current view is that neuron-to-neuron spreading of aS pathology contributes to the progression of a-synucleinopathy. We used an A53T mutant human aS transgenic mouse model (TgA53T) to examine whether the site of pathogenic aS inoculation affects the pattern of neuropathology and whether soluble and insoluble fractions derived from crude pathogenic tissue lysates exhibit differential capacities to initiate aS pathology. To test whether the inoculation site impacts the ultimate spatial/temporal patterns of aS pathology, aS preformed fibrils (PFF), or brain homogenates from TgA53T mice with a-synucleinopathy, were injected into the cortex/striatum, brain stem, or skeletal muscle. In all cases, inoculation of pathogenic aS induced end-stage motor dysfunction within ~100 days post-inoculation (dpi). Significantly, irrespective of the inoculation sites, ultimate distribution of the aS pathology was like that seen in normally aged TgA53T mice at end-stage, indicating that the intrinsic neuronal vulnerability is a significant determinant in the induction of aS pathology, even when initiated by inoculation of pathogenic aS. Temporal analysis of brain stem injected TgA53T mice show that initial aS pathology was seen by 30 days post-inoculation and inflammatory changes occur at later stages. To determine if the aS species with differential solubility are differentially pathogenic, brain lysates from end-stage TgA53Tmice were fractionated into highly soluble (S150) and insoluble (P150) fractions, as well as the endoplasmic reticulum (ER)-enriched fraction (P100). Significantly, all fractions were able to seed de novo aS pathology in vivo, when injected unilaterally into TgA53Tmice with the ER fractions being most pathogenic. Our results suggest that multiple aS species from brain can initiate the development of progressive aS pathology.
Collapse
|
14
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
15
|
Massey RS, Appadurai RR, Prakash R. A Surface Imprinted Polymer EIS Sensor for Detecting Alpha-Synuclein, a Parkinson's Disease Biomarker. MICROMACHINES 2024; 15:273. [PMID: 38399001 PMCID: PMC10892569 DOI: 10.3390/mi15020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Parkinson's Disease (PD) is a debilitating neurodegenerative disease, causing loss of motor function and, in some instances, cognitive decline and dementia in those affected. The quality of life can be improved, and disease progression delayed through early interventions. However, current methods of confirming a PD diagnosis are extremely invasive. This prevents their use as a screening tool for the early onset stages of PD. We propose a surface imprinted polymer (SIP) electroimpedance spectroscopy (EIS) biosensor for detecting α-Synuclein (αSyn) and its aggregates, a biomarker that appears in saliva and blood during the early stages of PD as the blood-brain barrier degrades. The surface imprinted polymer stamp is fabricated by low-temperature melt stamping polycaprolactone (PCL) on interdigitated EIS electrodes. The result is a low-cost, small-footprint biosensor that is highly suitable for non-invasive monitoring of the disease biomarker. The sensors were tested with αSyn dilutions in deionized water and in constant ionic concentration matrix solutions with decreasing concentrations of αSyn to remove the background effects of concentration. The device response confirmed the specificity of these devices to the target protein of monomeric αSyn. The sensor limit of detection was measured to be 5 pg/L, and its linear detection range was 5 pg/L-5 µg/L. This covers the physiological range of αSyn in saliva and makes this a highly promising method of quantifying αSyn monomers for PD patients in the future. The SIP surface was regenerated, and the sensor was reused to demonstrate its capability for repeat sensing as a potential continuous monitoring tool for the disease biomarker.
Collapse
Affiliation(s)
| | | | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; (R.S.M.); (R.R.A.)
| |
Collapse
|
16
|
Hong B, Ohtake Y, Itokazu T, Yamashita T. Glial senescence enhances α-synuclein pathology owing to its insufficient clearance caused by autophagy dysfunction. Cell Death Discov 2024; 10:50. [PMID: 38272865 PMCID: PMC10811334 DOI: 10.1038/s41420-024-01816-8] [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: 07/26/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Parkinson's disease (PD) is characterized by the pathological accumulation of α-synuclein (α-syn) and loss of dopaminergic neurons in the substantia nigra. Aging is a significant risk factor for PD. The accumulation of senescent glial cells in the aged brain contributes to PD progression by inducing chronic neuroinflammatory processes. However, although the insufficient degradation of α-syn aggregates results in PD deterioration, the possible alteration in the ability of α-syn clearance in senescent glia has received little attention. In this study, we investigated how aging and glial senescence affect the capacity of α-syn clearance. We found that following the intra-striatal injection of human α-syn (hu-α-syn) preformed fibril, hu-α-syn pathology persisted more in aged mice compared with younger mice and that aged microglia exhibited greater accumulation of hu-α-syn than younger microglia. Moreover, in vitro assay revealed that the clearance of hu-α-syn was primarily dependent on the autophagy-lysosome system rather than on the ubiquitin-proteasome system and that the capacity of hu-α-syn clearance was diminished in senescent glia because of autophagy-lysosome system dysfunction. Overall, this study provides new insights into the role of senescent glia in PD pathogenesis.
Collapse
Affiliation(s)
- Bin Hong
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yosuke Ohtake
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.
| |
Collapse
|
17
|
Radwan N, Khan E, Ardah MT, Kitada T, Haque ME. Ellagic Acid Prevents α-Synuclein Spread and Mitigates Toxicity by Enhancing Autophagic Flux in an Animal Model of Parkinson's Disease. Nutrients 2023; 16:85. [PMID: 38201915 PMCID: PMC10780534 DOI: 10.3390/nu16010085] [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: 11/11/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurological disorder, pathologically characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) as well as the formation of Lewy bodies composed mainly of α-synuclein (α-syn) aggregates. It has been documented that abnormal aggregation of α-syn is one of the major causes of developing PD. In the current study, administration of ellagic acid (EA), a polyphenolic compound (10 mg/kg bodyweight), significantly decreased α-syn spreading and preserved dopaminergic neurons in a male C57BL/6 mouse model of PD. Moreover, EA altered the autophagic flux, suggesting the involvement of a restorative mechanism meditated by EA treatment. Our data support that EA could play a major role in the clearing of toxic α-syn from spreading, in addition to the canonical antioxidative role, and thus preventing dopaminergic neuronal death.
Collapse
Affiliation(s)
- Nada Radwan
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (N.R.); (E.K.); (M.T.A.)
| | - Engila Khan
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (N.R.); (E.K.); (M.T.A.)
| | - Mustafa T. Ardah
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (N.R.); (E.K.); (M.T.A.)
| | - Tohru Kitada
- Otawa-Kagaku, Parkinson Clinic and Research, Kamakura 247-0061, Japan;
| | - M. Emdadul Haque
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (N.R.); (E.K.); (M.T.A.)
| |
Collapse
|
18
|
Park JS, Ahmad R, Choe K, Kang MH, Park TJ, Kim MO. Immunization Effects of a Novel α-Synuclein-Based Peptide Epitope Vaccine in Parkinson's Disease-Associated Pathology. Vaccines (Basel) 2023; 11:1820. [PMID: 38140224 PMCID: PMC10748214 DOI: 10.3390/vaccines11121820] [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: 09/29/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease that affects the central nervous system, specifically the motor system. It is mainly caused by the loss of dopamine due to the accumulation of α-synuclein (α-syn) protein in the striatum and substantia nigra pars compacta (SNpc). Previous studies have reported that immunization may be a potential preventive strategy for neurodegenerative diseases such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Therefore, the aim of the study was to design an α-syn specific epitope vaccine and investigate its effect in PD-related pathophysiology using an α-syn-induced mouse model. We used an in silico model to identify and design a non-toxic α-syn-based peptide epitope vaccine and, to overcome poor immunogenicity, the vaccine was coupled with immunogenic carrier proteins, i.e., ovalbumin (OVA) and keyhole limpet haemocyanin (KLH). Our results showed that vaccinated PD mouse models, especially with vaccines with carrier proteins, improved in motor functions compared with the non-vaccinated PD model. Additionally, the vaccinated groups showed increased immunoglobulin G (IgG) levels in the spleen and plasma as well as decreased interleukin-10 (IL-10) levels in the plasma. Furthermore, vaccinated groups, especially OVA and KLH groups, showed decrease in α-syn levels and increased dopamine-related markers, i.e., tyrosine hydroxylase (TH), vesicle monoamine transporter 2 (VMAT2), and dopamine transporter (DAT), and autophagy activities in the striatum and SNpc. Lastly, our data showed decreased neuroinflammation by reducing the activation of microglia and astrocytes and pro-inflammatory cytokines in the immunized groups, especially with OVA and KLH carrier proteins. Overall, these results suggest that vaccination, especially with immunogenic carrier proteins, is effective in reducing the accumulation of α-syn aggregates in the brain and ameliorate PD-related pathophysiology. Hence, further development of this approach might have a potential role in preventing the development of PD.
Collapse
Affiliation(s)
- Jun Sung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.S.P.); (R.A.); (M.H.K.)
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.S.P.); (R.A.); (M.H.K.)
| | - Kyonghwan Choe
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.S.P.); (R.A.); (M.H.K.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Min Hwa Kang
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.S.P.); (R.A.); (M.H.K.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, UK;
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.S.P.); (R.A.); (M.H.K.)
- Alz-Dementia Korea Co., Jinju 52828, Republic of Korea
| |
Collapse
|
19
|
Teil M, Dovero S, Bourdenx M, Arotcarena ML, Darricau M, Porras G, Thiolat ML, Trigo-Damas I, Perier C, Estrada C, Garcia-Carrillo N, Herrero MT, Vila M, Obeso JA, Bezard E, Dehay B. Cortical Lewy body injections induce long-distance pathogenic alterations in the non-human primate brain. NPJ Parkinsons Dis 2023; 9:135. [PMID: 37726343 PMCID: PMC10509171 DOI: 10.1038/s41531-023-00579-w] [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: 05/05/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
Aggregation of α-synuclein (α-syn) is the cornerstone of neurodegenerative diseases termed synucleinopathies, which include Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), and Multiple System Atrophy (MSA). These synucleinopathies are characterized by the deposit of aggregated α-syn in intracellular inclusions observable in neurons and glial cells. In PD and DLB, these aggregates, predominantly located in neurons, are called Lewy Bodies (LBs). These LBs are one of the pathological hallmarks of PD and DLB, alongside dopaminergic neuron loss in the substantia nigra. Previous studies have demonstrated the ability of PD patient-derived LB fractions to induce nigrostriatal neurodegeneration and α-syn pathology when injected into the striatum or the enteric nervous system of non-human primates. Here, we report the pathological consequences of injecting these LB fractions into the cortex of non-human primates. To this end, we inoculated mesencephalic PD patient-derived LB fractions into the prefrontal cortex of baboon monkeys terminated one year later. Extensive analyses were performed to evaluate pathological markers known to be affected in LB pathologies. We first assessed the hypothesized presence of phosphorylated α-syn at S129 (pSyn) in the prefrontal cortices. Second, we quantified the neuronal, microglial, and astrocytic cell survival in the same cortices. Third, we characterized these cortical LB injections' putative impact on the integrity of the nigrostriatal system. Overall, we observed pSyn accumulation around the injection site in the dorsal prefrontal cortex, in connected cortical regions, and further towards the striatum, suggesting α-syn pathological propagation. The pathology was also accompanied by neuronal loss in these prefrontal cortical regions and the caudate nucleus, without, however, loss of nigral dopamine neurons. In conclusion, this pilot study provides novel data demonstrating the toxicity of patient-derived extracts, their potential to propagate from the cortex to the striatum in non-human primates, and a possible primate model of DLB.
Collapse
Affiliation(s)
- Margaux Teil
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | - Sandra Dovero
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | - Mathieu Bourdenx
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | | | | | - Gregory Porras
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | | | - Inés Trigo-Damas
- HM CINAC, HM Puerta del Sur, Fundación HM Hospitales and CIBERNED and CEU-San Pablo University Madrid, E-28938, Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Celine Perier
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Cristina Estrada
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100, Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100, Murcia, Spain
| | - Nuria Garcia-Carrillo
- Centro Experimental en Investigaciones Biomédica (CEIB), Universidad de Murcia, Murcia, Spain
| | - María Trinidad Herrero
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100, Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100, Murcia, Spain
| | - Miquel Vila
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - José A Obeso
- HM CINAC, HM Puerta del Sur, Fundación HM Hospitales and CIBERNED and CEU-San Pablo University Madrid, E-28938, Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- CEU, San Pablo University Madrid, E-28938 Mostoles, Spain 2 HM CINAC, HM Puerta del Sur and CIBERNED and CEU-San Pablo University Madrid, E-, 28938, Mostoles, Spain
| | - Erwan Bezard
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France.
| |
Collapse
|
20
|
Ofori K, Ghosh A, Verma DK, Wheeler D, Cabrera G, Seo JB, Kim YH. A Novel NOX Inhibitor Alleviates Parkinson's Disease Pathology in PFF-Injected Mice. Int J Mol Sci 2023; 24:14278. [PMID: 37762579 PMCID: PMC10531511 DOI: 10.3390/ijms241814278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress-mediated damage is often a downstream result of Parkinson's disease (PD), which is marked by sharp decline in dopaminergic neurons within the nigrostriatal regions of the brain, accounting for the symptomatic motor deficits in patients. Regulating the level of oxidative stress may present a beneficial approach in preventing PD pathology. Here, we assessed the efficacy of a nicotinamide adenine phosphate (NADPH) oxidase (NOX) inhibitor, an exogenous reactive oxygen species (ROS) regulator synthesized by Aptabio therapeutics with the specificity to NOX-1, 2 and 4. Utilizing N27 rat dopaminergic cells and C57Bl/6 mice, we confirmed that the exposures of alpha-synuclein preformed fibrils (PFF) induced protein aggregation, a hallmark in PD pathology. In vitro assessment of the novel compound revealed an increase in cell viability and decreases in cytotoxicity, ROS, and protein aggregation (Thioflavin-T stain) against PFF exposure at the optimal concentration of 10 nM. Concomitantly, the oral treatment alleviated motor-deficits in behavioral tests, such as hindlimb clasping, rotarod, pole, nesting and grooming test, via reducing protein aggregation, based on rescued dopaminergic neuronal loss. The suppression of NOX-1, 2 and 4 within the striatum and ventral midbrain regions including Substantia Nigra compacta (SNc) contributed to neuroprotective/recovery effects, making it a potential therapeutic option for PD.
Collapse
Affiliation(s)
- Kwadwo Ofori
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Anurupa Ghosh
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Dinesh Kumar Verma
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Darice Wheeler
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Gabriela Cabrera
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Jong-Bok Seo
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Yong-Hwan Kim
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| |
Collapse
|
21
|
Zhang S, Dauer K, Strohäker T, Tatenhorst L, Caldi Gomes L, Mayer S, Jung BC, Kim WS, Lee S, Becker S, Liesche‐Starnecker F, Zweckstetter M, Lingor P. Alpha-synuclein fibrils amplified from multiple system atrophy and Parkinson's disease patient brain spread after intracerebral injection into mouse brain. Brain Pathol 2023; 33:e13196. [PMID: 37485772 PMCID: PMC10467043 DOI: 10.1111/bpa.13196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB) are neurodegenerative disorders with alpha-synuclein (α-syn) aggregation pathology. Different strains of α-syn with unique properties are suggested to cause distinct clinical and pathological manifestations resulting in PD, MSA, or DLB. To study individual α-syn spreading patterns, we injected α-syn fibrils amplified from brain homogenates of two MSA patients and two PD patients into the brains of C57BI6/J mice. Antibody staining against pS129-α-syn showed that α-syn fibrils amplified from the brain homogenates of the four different patients caused different levels of α-syn spreading. The strongest α-syn pathology was triggered by α-syn fibrils of one of the two MSA patients, followed by comparable pS129-α-syn induction by the second MSA and one PD patient material. Histological analysis using an antibody against Iba1 further showed that the formation of pS129-α-syn is associated with increased microglia activation. In contrast, no differences in dopaminergic neuron numbers or co-localization of α-syn in oligodendrocytes were observed between the different groups. Our data support the spreading of α-syn pathology in MSA, while at the same time pointing to spreading heterogeneity between different patients potentially driven by individual patient immanent factors.
Collapse
Affiliation(s)
- Shuyu Zhang
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Karina Dauer
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Timo Strohäker
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Lars Tatenhorst
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Lucas Caldi Gomes
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Simon Mayer
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Byung Chul Jung
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Woojin S. Kim
- Faculty of Medicine and Health, Brain and Mind Centre and School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesUniversity of New South Wales and Neuroscience Research AustraliaRandwickNew South WalesAustralia
| | - Seung‐Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Stefan Becker
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Friederike Liesche‐Starnecker
- Department of Neuropathology, Institute of Pathology, School of MedicineTechnical University MunichMunichGermany
- Department of Pathology and Molecular Diagnostics, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Paul Lingor
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
22
|
Massey RS, McConnell EM, Chan D, Holahan MR, DeRosa MC, Prakash R. Non-invasive Monitoring of α-Synuclein in Saliva for Parkinson's Disease Using Organic Electrolyte-Gated FET Aptasensor. ACS Sens 2023; 8:3116-3126. [PMID: 37506391 DOI: 10.1021/acssensors.3c00757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Parkinson's disease (PD) currently affects more than 1 million people in the US alone, with nearly 8.5 million suffering from the disease worldwide, as per the World Health Organization. However, there remains no fast, pain-free, and effective method of screening for the disease in the ageing population, which also happens to be the most susceptible to this neurodegenerative disease. αSynuclein (αSyn) is a promising PD biomarker, demonstrating clear delineations between levels of the αSyn monomer and the extent of αSyn aggregation in the saliva of PD patients and healthy controls. In this work, we have demonstrated a laboratory prototype of a soft fluidics integrated organic electrolyte-gated field-effect transistor (OEGFET) aptasensor platform capable of quantifying levels of αSyn aggregation in saliva. The aptasensor relies on a recently reported synthetic aptamer which selectively binds to αSyn monomer as the bio-recognition molecule within the integrated fluidic channel of the biosensor. The produced saliva sensor is label-free, fast, and reusable, demonstrating good selectivity only to the target molecule in its monomer form. The novelty of these devices is the fully isolated organic semiconductor, which extends the shelf life, and the novel fully integrated soft microfluidic channels, which simplify saliva loading and testing. The OEGFET aptasensor has a limit of detection of 10 fg/L for the αSyn monomer in spiked saliva supernatant solutions, with a linear range of 100 fg/L to 10 μg/L. The linear range covers the physiological range of the αSyn monomer in the saliva of PD patients. Our biosensors demonstrate a desirably low limit of detection, an extended linear range, and fully integrated microchannels for saliva sample handling, making them a promising platform for non-invasive point-of-care testing of PD.
Collapse
Affiliation(s)
- Roslyn S Massey
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S5B6, Canada
| | - Erin M McConnell
- Department of Chemistry and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Dennis Chan
- Dept of Neuroscience, Health Sciences Building, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Matthew R Holahan
- Dept of Neuroscience, Health Sciences Building, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Maria C DeRosa
- Department of Chemistry and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
23
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
24
|
Gorecki AM, Spencer H, Meloni BP, Anderton RS. The Poly-Arginine Peptide R18D Interferes with the Internalisation of α-Synuclein Pre-Formed Fibrils in STC-1 Enteroendocrine Cells. Biomedicines 2023; 11:2089. [PMID: 37626586 PMCID: PMC10452853 DOI: 10.3390/biomedicines11082089] [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: 07/06/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
In Parkinson's disease (PD), gut inflammation is hypothesised to contribute to α-synuclein aggregation, but gastrointestinal α-synuclein expression is poorly characterised. Cationic arginine-rich peptides (CARPs) are an emerging therapeutic option that exerts various neuroprotective effects and may target the transmission of protein aggregates. This study aimed to investigate endogenous α-synuclein expression in enteroendocrine STC-1 cells and the potential of the CARP, R18D (18-mer of D-arginine), to prevent internalisation of pre-formed α-synuclein fibrils (PFFs) in enteroendocrine cells in vitro. Through confocal microscopy, the immunoreactivity of full-length α-synuclein and the serine-129 phosphorylated form (pS129) was investigated in STC-1 (mouse enteroendocrine) cells. Thereafter, STC-1 cells were exposed to PFFs tagged with Alexa-Fluor 488 (PFF-488) for 2 and 24 h and R18D-FITC for 10 min. After confirming the uptake of both PFFs and R18D-FITC through fluorescent microscopy, STC-1 cells were pre-treated with R18D (5 or 10 μM) for 10 min prior to 2 h of PFF-488 exposure. Immunoreactivity for endogenous α-synuclein and pS129 was evident in STC-1 cells, with prominent pS129 staining along cytoplasmic processes and in perinuclear areas. STC-1 cells internalised PFFs, confirmed through co-localisation of PFF-488 and human-specific α-synuclein immunoreactivity. R18D-FITC entered STC-1 cells within 10 min and pre-treatment of STC-1 cells with R18D interfered with PFF uptake. The endogenous presence of α-synuclein in enteroendocrine cells, coupled with their rapid uptake of PFFs, demonstrates a potential for pathogenic spread of α-synuclein aggregates in the gut. R18D is a novel therapeutic approach to reduce the intercellular transmission of α-synuclein pathology.
Collapse
Affiliation(s)
- Anastazja M. Gorecki
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (H.S.)
- School of Biological Sciences, University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Holly Spencer
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (H.S.)
| | - Bruno P. Meloni
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, First Floor, G-Block, QEII Medical Centre, Nedlands, WA 6008, Australia
| | - Ryan S. Anderton
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (H.S.)
| |
Collapse
|
25
|
Dovonou A, Bolduc C, Soto Linan V, Gora C, Peralta Iii MR, Lévesque M. Animal models of Parkinson's disease: bridging the gap between disease hallmarks and research questions. Transl Neurodegener 2023; 12:36. [PMID: 37468944 PMCID: PMC10354932 DOI: 10.1186/s40035-023-00368-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still provide valuable information that contributes to our understanding of the disease and the limitations of our treatment options. This review comprehensively summarizes the different animal models available for Parkinson's research, with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to realize that the strengths and weaknesses of each model and the induction technique at our disposal are determined by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete discernment of classical and novel animal models in PD research.
Collapse
Affiliation(s)
- Axelle Dovonou
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Cyril Bolduc
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Victoria Soto Linan
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Charles Gora
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Modesto R Peralta Iii
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
26
|
Mirzac D, Kreis SL, Luhmann HJ, Gonzalez-Escamilla G, Groppa S. Translating Pathological Brain Activity Primers in Parkinson's Disease Research. RESEARCH (WASHINGTON, D.C.) 2023; 6:0183. [PMID: 37383218 PMCID: PMC10298229 DOI: 10.34133/research.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Translational experimental approaches that help us better trace Parkinson's disease (PD) pathophysiological mechanisms leading to new therapeutic targets are urgently needed. In this article, we review recent experimental and clinical studies addressing abnormal neuronal activity and pathological network oscillations, as well as their underlying mechanisms and modulation. Our aim is to enhance our knowledge about the progression of Parkinson's disease pathology and the timing of its symptom's manifestation. Here, we present mechanistic insights relevant for the generation of aberrant oscillatory activity within the cortico-basal ganglia circuits. We summarize recent achievements extrapolated from available PD animal models, discuss their advantages and limitations, debate on their differential applicability, and suggest approaches for transferring knowledge on disease pathology into future research and clinical applications.
Collapse
Affiliation(s)
- Daniela Mirzac
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Svenja L. Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sergiu Groppa
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
27
|
Béreau M, Van Waes V, Servant M, Magnin E, Tatu L, Anheim M. Apathy in Parkinson's Disease: Clinical Patterns and Neurobiological Basis. Cells 2023; 12:1599. [PMID: 37371068 DOI: 10.3390/cells12121599] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Apathy is commonly defined as a loss of motivation leading to a reduction in goal-directed behaviors. This multidimensional syndrome, which includes cognitive, emotional and behavioral components, is one of the most prevalent neuropsychiatric features of Parkinson's disease (PD). It has been established that the prevalence of apathy increases as PD progresses. However, the pathophysiology and anatomic substrate of this syndrome remain unclear. Apathy seems to be underpinned by impaired anatomical structures that link the prefrontal cortex with the limbic system. It can be encountered in the prodromal stage of the disease and in fluctuating PD patients receiving bilateral chronic subthalamic nucleus stimulation. In these stages, apathy may be considered as a disorder of motivation that embodies amotivational behavioral syndrome, is underpinned by combined dopaminergic and serotonergic denervation and is dopa-responsive. In contrast, in advanced PD patients, apathy may be considered as cognitive apathy that announces cognitive decline and PD dementia, is underpinned by diffuse neurotransmitter system dysfunction and Lewy pathology spreading and is no longer dopa-responsive. In this review, we discuss the clinical patterns of apathy and their treatment, the neurobiological basis of apathy, the potential role of the anatomical structures involved and the pathways in motivational and cognitive apathy.
Collapse
Affiliation(s)
- Matthieu Béreau
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Vincent Van Waes
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Mathieu Servant
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Eloi Magnin
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
| | - Laurent Tatu
- Département de Neurologie, CHU de Besançon, 25000 Besançon, France
- Université de Franche-Comté, LINC Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, 25000 Besançon, France
- Laboratoire d'Anatomie, Université de Franche-Comté, 25000 Besançon, France
| | - Mathieu Anheim
- Département de Neurologie, CHU de Strasbourg, 67200 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
- Institut de génétique Et de Biologie Moléculaire Et Cellulaire (IGBMC), INSERM-U964, CNRS-UMR7104, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
28
|
Pérez-Acuña D, Rhee KH, Shin SJ, Ahn J, Lee JY, Lee SJ. Retina-to-brain spreading of α-synuclein after intravitreal injection of preformed fibrils. Acta Neuropathol Commun 2023; 11:83. [PMID: 37210559 PMCID: PMC10199563 DOI: 10.1186/s40478-023-01575-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/22/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the aggregation of misfolded α-synuclein and progressive spreading of the aggregates from a few discrete regions to wider brain regions. Although PD has been classically considered a movement disorder, a large body of clinical evidence has revealed the progressive occurrence of non-motor symptoms. Patients present visual symptoms in the initial stages of the disease, and accumulation of phospho-α-synuclein, dopaminergic neuronal loss, and retinal thinning has been observed in the retinas of PD patients. Based on such human data, we hypothesized that α-synuclein aggregation can initiate in the retina and spread to the brain through the visual pathway. Here, we demonstrate accumulation of α-synuclein in the retinas and brains of naive mice after intravitreal injection of α-synuclein preformed fibrils (PFFs). Histological analyses showed deposition of phospho-α-synuclein inclusions within the retina 2 months after injection, with increased oxidative stress leading to loss of retinal ganglion cells and dopaminergic dysfunction. In addition, we found accumulation of phospho-α-synuclein in cortical areas with accompanying neuroinflammation after 5 months. Collectively, our findings suggest that retinal synucleinopathy lesions initiated by intravitreal injection of α-synuclein PFFs spread to various brain regions through the visual pathway in mice.
Collapse
Affiliation(s)
- Dayana Pérez-Acuña
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
| | - Ka Hyun Rhee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Soo Jean Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
| | - Jeeyun Ahn
- Department of Ophthalmology, College of Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University, Seoul, South Korea
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, South Korea.
- Neuramedy, Seoul, South Korea.
| |
Collapse
|
29
|
Emmi A, Sandre M, Russo FP, Tombesi G, Garrì F, Campagnolo M, Carecchio M, Biundo R, Spolverato G, Macchi V, Savarino E, Farinati F, Parchi P, Porzionato A, Bubacco L, De Caro R, Kovacs GG, Antonini A. Duodenal alpha-Synuclein Pathology and Enteric Gliosis in Advanced Parkinson's Disease. Mov Disord 2023; 38:885-894. [PMID: 36847308 DOI: 10.1002/mds.29358] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND The role of the gut-brain axis has been recently highlighted as a major contributor to Parkinson's disease (PD) physiopathology, with numerous studies investigating bidirectional transmission of pathological protein aggregates, such as α-synuclein (αSyn). However, the extent and the characteristics of pathology in the enteric nervous system have not been fully investigated. OBJECTIVE We characterized αSyn alterations and glial responses in duodenum biopsies of patients with PD by employing topography-specific sampling and conformation-specific αSyn antibodies. METHODS We examined 18 patients with advanced PD who underwent Duodopa percutaneous endoscopic gastrostomy and jejunal tube procedure, 4 untreated patients with early PD (disease duration <5 years), and 18 age- and -sex-matched healthy control subjects undergoing routine diagnostic endoscopy. A mean of four duodenal wall biopsies were sampled from each patient. Immunohistochemistry was performed for anti-aggregated αSyn (5G4) and glial fibrillary acidic protein antibodies. Morphometrical semiquantitative analysis was performed to characterize αSyn-5G4+ and glial fibrillary acidic protein-positive density and size. RESULTS Immunoreactivity for aggregated α-Syn was identified in all patients with PD (early and advanced) compared with controls. αSyn-5G4+ colocalized with neuronal marker β-III-tubulin. Evaluation of enteric glial cells demonstrated an increased size and density when compared with controls, suggesting reactive gliosis. CONCLUSIONS We found evidence of synuclein pathology and gliosis in the duodenum of patients with PD, including early de novo cases. Future studies are required to evaluate how early in the disease process duodenal pathology occurs and its possible contribution to levodopa effect in chronic patients. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aron Emmi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Michele Sandre
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Francesco Paolo Russo
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Giulia Tombesi
- Department of Biology, University of Padova, Padova, Italy
| | - Federica Garrì
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
| | - Marta Campagnolo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Roberta Biundo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of General Psychology, University of Padova, Padova, Italy
| | - Gaya Spolverato
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Veronica Macchi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Edoardo Savarino
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Fabio Farinati
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Piero Parchi
- Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, Ontario, Canada
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Porzionato
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Luigi Bubacco
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Departments of Laboratory Medicine and Pathobiology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine Program & Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Padua Neuroscience Center (PNC), Department of Neuroscience, University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
| |
Collapse
|
30
|
Guo Q, Kawahata I, Jia W, Wang H, Cheng A, Yabuki Y, Shioda N, Fukunaga K. α-Synuclein decoy peptide protects mice against α-synuclein-induced memory loss. CNS Neurosci Ther 2023; 29:1547-1560. [PMID: 36786129 PMCID: PMC10173724 DOI: 10.1111/cns.14120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
AIMS We previously found that a decoy peptide derived from the C-terminal sequence of α-Synuclein (αSyn) prevents cytotoxic αSyn aggregation caused by fatty acid-binding protein 3 (FABP3) in vitro. In this study, we continued to utilize αSyn-derived peptides to further validate their effects on αSyn neurotoxicity and behavioral impairments in αSyn preformed fibrils (PFFs)-injected mouse model of Parkinson's disease (PD). METHODS Mice were injected with αSyn PFFs in the bilateral olfactory bulb (OB) and then were subjected to behavioral analysis at 2-week intervals post-injection. Peptides nasal administration was initiated one week after injection. Changes in phosphorylation of αSyn and neuronal damage in the OB were measured using immunostaining at week 4. The effect of peptides on the interaction between αSyn and FABP3 was examined using co-immunoprecipitation. RESULTS αSyn PFF-injected mice showed significant memory loss but no motor function impairment. Long-term nasal treatment with peptides effectively prevented memory impairment. In peptide-treated αSyn PFF-injected mice, the peptides entered the OB smoothly through the nasal cavity and were mainly concentrated in neurons in the mitral cell layer, significantly suppressing the excessive phosphorylation of αSyn and reducing the formation of αSyn-FABP3 oligomers, thereby preventing neuronal death. The addition of peptides also blocked the interaction of αSyn and FABP3 at the recombinant protein level, and its effect was strongest at molar concentrations comparable to those of αSyn and FABP3. CONCLUSIONS Our findings suggest that the αSyn decoy peptide represents a novel therapeutic approach for reducing the accumulation of toxic αSyn-FABP3 oligomers in the brain, thereby preventing the progression of synucleinopathies.
Collapse
Affiliation(s)
- Qingyun Guo
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wenbin Jia
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haoyang Wang
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - An Cheng
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.,BRI Pharma Incorporated, Sendai, Japan
| |
Collapse
|
31
|
The Role of α-Synuclein in the Regulation of Serotonin System: Physiological and Pathological Features. Biomedicines 2023; 11:biomedicines11020541. [PMID: 36831077 PMCID: PMC9953742 DOI: 10.3390/biomedicines11020541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
In patients affected by Parkinson's disease (PD), up to 50% of them experience cognitive changes, and psychiatric disturbances, such as anxiety and depression, often precede the onset of motor symptoms and have a negative impact on their quality of life. Pathologically, PD is characterized by the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of intracellular inclusions, called Lewy bodies and Lewy neurites, composed mostly of α-synuclein (α-Syn). Much of PD research has focused on the role of α-Syn aggregates in the degeneration of SNc DA neurons due to the impact of striatal DA deficits on classical motor phenotypes. However, abundant Lewy pathology is also found in other brain regions including the midbrain raphe nuclei, which may contribute to non-motor symptoms. Indeed, dysfunction of the serotonergic (5-HT) system, which regulates mood and emotional pathways, occurs during the premotor phase of PD. However, little is known about the functional consequences of α-Syn inclusions in this neuronal population other than DA neurons. Here, we provide an overview of the current knowledge of α-Syn and its role in regulating the 5-HT function in health and disease. Understanding the relative contributions to α-Syn-linked alterations in the 5-HT system may provide a basis for identifying PD patients at risk for developing depression and could lead to a more targeted therapeutic approach.
Collapse
|
32
|
Herman S, Djaldetti R, Mollenhauer B, Offen D. CSF-derived extracellular vesicles from patients with Parkinson's disease induce symptoms and pathology. Brain 2023; 146:209-224. [PMID: 35881523 DOI: 10.1093/brain/awac261] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease is characterized by the gradual appearance of intraneuronal inclusions that are primarily composed of misfolded α-synuclein protein, leading to cytotoxicity and neural death. Recent in vitro and in vivo studies suggest that misfolded α-synuclein may spread transcellularly in a prion-like manner, inducing pathological aggregates in healthy neurons, and is disseminated via secretion of extracellular vesicles. Accordingly, extracellular vesicles derived from brain lysates and CSF of patients with Parkinson's disease were shown to facilitate α-synuclein aggregation in healthy cells. Prompted by the hypothesis of Braak and colleagues that the olfactory bulb is one of the primary propagation sites for the initiation of Parkinson's disease, we sought to investigate the role of extracellular vesicles in the spread of α-synuclein and progression of Parkinson's disease through the olfactory bulb. Extracellular vesicles derived from the CSF of patients diagnosed with Parkinson's disease or with a non-synucleinopathy neurodegenerative disorder were administered intranasally to healthy mice, once daily over 4 days. Three months later, mice were subjected to motor and non-motor tests. Functional impairments were elucidated by histochemical analysis of midbrain structures relevant to Parkinson's disease pathology, 8 months after EVs treatment. Mice treated with extracellular vesicles from the patients with Parkinson's disease displayed multiple symptoms consistent with prodromal and clinical-phase Parkinson's disease such as hyposmia, motor behaviour impairments and high anxiety levels. Furthermore, their midbrains showed widespread α-synuclein aggregations, dopaminergic neurodegeneration, neuroinflammation and altered autophagy activity. Several unconventional pathologies were also observed, such as α-synuclein aggregations in the red nucleus, growth of premature grey hair and astrogliosis. Collectively, these data indicate that intranasally administered extracellular vesicles derived from the CSF of patients with Parkinson's disease can propagate α-synuclein aggregation in vivo and trigger Parkinson's disease-like symptoms and pathology in healthy mice.
Collapse
Affiliation(s)
- Shay Herman
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ruth Djaldetti
- Department of Neurology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Paracelsus-Elena-Klinik, Kassel, Germany
| | - Daniel Offen
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
33
|
Pinto-Costa R, Harbachova E, La Vitola P, Di Monte DA. Overexpression-Induced α-Synuclein Brain Spreading. Neurotherapeutics 2023; 20:83-96. [PMID: 36512255 PMCID: PMC10119350 DOI: 10.1007/s13311-022-01332-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
Interneuronal transfer of pathological α-synuclein species is thought to play an important role in the progressive advancement of Lewy pathology and increasing severity of clinical manifestations in Parkinson's and other diseases commonly referred to as synucleinopathies. Pathophysiological conditions and mechanisms triggering this trans-synaptic spreading bear therefore significant pathogenetic implications but have yet to be fully elucidated. In vivo experimental models support the conclusion that increased expression of intraneuronal α-synuclein can itself induce protein spreading throughout the brain as well as from the brain to peripheral tissues. For example, overexpression of α-synuclein targeted to the rodent dorsal medulla oblongata results in its transfer and accumulation into recipient axons innervating this brain region; through these axons, α-synuclein can then travel caudo-rostrally and reach other brain sites in the pons, midbrain, and forebrain. When protein overexpression is induced in the rodent midbrain, long-distance α-synuclein spreading can be followed over time; spreading-induced α-synuclein accumulation affects lower brain regions, including the dorsal motor nucleus of the vagus, proceeds through efferent axons of the vagus nerve, and is ultimately detected within vagal motor nerve endings in the gastric wall. As discussed in this review, animal models featuring α-synuclein overexpression not only support a relationship between α-synuclein burden and protein spreading but have also provided important clues on conditions/mechanisms capable of promoting interneuronal α-synuclein transfer. Intriguing findings include the relationship between neuronal activity and protein spreading and the role of oxidant stress in trans-synaptic α-synuclein mobility.
Collapse
Affiliation(s)
- Rita Pinto-Costa
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Eugenia Harbachova
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Pietro La Vitola
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Donato A Di Monte
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany.
| |
Collapse
|
34
|
Maharjan N, Saxena S. Models of Neurodegenerative Diseases. Neurogenetics 2023. [DOI: 10.1007/978-3-031-07793-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Shin JW, An S, Kim D, Kim H, Ahn J, Eom J, You WK, Yun H, Lee B, Sung B, Jung J, Kim S, Son Y, Sung E, Lee H, Lee S, Song D, Pak Y, Sandhu JK, Haqqani AS, Stanimirovic DB, Yoo J, Kim D, Maeng S, Lee J, Lee SH. Grabody B, an IGF1 receptor-based shuttle, mediates efficient delivery of biologics across the blood-brain barrier. CELL REPORTS METHODS 2022; 2:100338. [PMID: 36452865 PMCID: PMC9701613 DOI: 10.1016/j.crmeth.2022.100338] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/26/2021] [Accepted: 10/21/2022] [Indexed: 06/17/2023]
Abstract
Effective delivery of therapeutics to the brain is challenging. Molecular shuttles use receptors expressed on brain endothelial cells to deliver therapeutics. Antibodies targeting transferrin receptor (TfR) have been widely developed as molecular shuttles. However, the TfR-based approach raises concerns about safety and developmental burden. Here, we report insulin-like growth factor 1 receptor (IGF1R) as an ideal target for the molecular shuttle. We also describe Grabody B, an antibody against IGF1R, as a molecular shuttle. Grabody B has broad cross-species reactivity and does not interfere with IGF1R-mediated signaling. We demonstrate that administration of Grabody B-fused anti-alpha-synuclein (α-Syn) antibody induces better improvement in neuropathology and behavior in a Parkinson's disease animal model than the therapeutic antibody alone due to its superior serum pharmacokinetics and enhanced brain exposure. The results indicate that IGF1R is an ideal shuttle target and Grabody B is a safe and efficient molecular shuttle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hyesu Yun
- ABL Bio, Inc., Seongnam-si, South Korea
| | - Bora Lee
- ABL Bio, Inc., Seongnam-si, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sungho Maeng
- Department of Comprehensive Health Science, Kyung Hee University, Yongin-si, South Korea
| | - Jeonghun Lee
- Department of Comprehensive Health Science, Kyung Hee University, Yongin-si, South Korea
| | | |
Collapse
|
36
|
Padilla-Godínez FJ, Ruiz-Ortega LI, Guerra-Crespo M. Nanomedicine in the Face of Parkinson's Disease: From Drug Delivery Systems to Nanozymes. Cells 2022; 11:3445. [PMID: 36359841 PMCID: PMC9657131 DOI: 10.3390/cells11213445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 01/02/2024] Open
Abstract
The complexity and overall burden of Parkinson's disease (PD) require new pharmacological approaches to counteract the symptomatology while reducing the progressive neurodegeneration of affected dopaminergic neurons. Since the pathophysiological signature of PD is characterized by the loss of physiological levels of dopamine (DA) and the misfolding and aggregation of the alpha-synuclein (α-syn) protein, new proposals seek to restore the lost DA and inhibit the progressive damage derived from pathological α-syn and its impact in terms of oxidative stress. In this line, nanomedicine (the medical application of nanotechnology) has achieved significant advances in the development of nanocarriers capable of transporting and delivering basal state DA in a controlled manner in the tissues of interest, as well as highly selective catalytic nanostructures with enzyme-like properties for the elimination of reactive oxygen species (responsible for oxidative stress) and the proteolysis of misfolded proteins. Although some of these proposals remain in their early stages, the deepening of our knowledge concerning the pathological processes of PD and the advances in nanomedicine could endow for the development of potential treatments for this still incurable condition. Therefore, in this paper, we offer: (i) a brief summary of the most recent findings concerning the physiology of motor regulation and (ii) the molecular neuropathological processes associated with PD, together with (iii) a recapitulation of the current progress in controlled DA release by nanocarriers and (iv) the design of nanozymes, catalytic nanostructures with oxidoreductase-, chaperon, and protease-like properties. Finally, we conclude by describing the prospects and knowledge gaps to overcome and consider as research into nanotherapies for PD continues, especially when clinical translations take place.
Collapse
Affiliation(s)
- Francisco J. Padilla-Godínez
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| | - Leonardo I. Ruiz-Ortega
- Institute for Physical Sciences, National Autonomous University of Mexico, Cuernavaca 62210, Mexico
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Magdalena Guerra-Crespo
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| |
Collapse
|
37
|
Song MK, Adams L, Lee JH, Kim YS. NXP031 prevents dopaminergic neuronal loss and oxidative damage in the AAV-WT-α-synuclein mouse model of Parkinson’s disease. PLoS One 2022; 17:e0272085. [PMID: 35901090 PMCID: PMC9333296 DOI: 10.1371/journal.pone.0272085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/12/2022] [Indexed: 11/18/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by inclusions of aggregated α-synuclein (α-Syn). Oxidative stress plays a critical role in nigrostriatal degeneration and is responsible for α-Syn aggregation in PD. Vitamin C or ascorbic acid acts as an effective antioxidant to prevent free radical damage. However, vitamin C is easily oxidized and often loses its physiological activity, limiting its therapeutic potential. The objective of this study was to evaluate whether NXP031, a new compound we developed consisting of Aptamin C and Vitamin C, is neuroprotective against α-synucleinopathy. To model α-Syn induced PD, we stereotactically injected AAV particles overexpressing human α-Syn into the substantia nigra (SN) of mice. One week after AAV injection, NXP031 was administered via oral gavage every day for eight weeks. We found that oral administration of NXP031 ameliorated motor deficits measured by the rotarod test and prevented the loss of nigral dopaminergic neurons caused by WT-α-Syn overexpression in the SN. Also, NXP031 blocked the propagation of aggregated α-Syn into the hippocampus by alleviating oxidative stress. These results indicate that NXP031 can be a potential therapeutic for PD.
Collapse
Affiliation(s)
- Min Kyung Song
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, United States of America
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Levi Adams
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, United States of America
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Joo Hee Lee
- College of Nursing Science, Kyung Hee University, Seoul, Republic of Korea
| | - Yoon-Seong Kim
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, United States of America
- Burnett School of Biomedical Sciences, UCF College of Medicine, University of Central Florida, Orlando, FL, United States of America
- Nexmos Co Ltd, Yongin-Si, Gyeonggi-Do, Republic of Korea
- * E-mail:
| |
Collapse
|
38
|
Ghosh AA, Verma DK, Cabrera G, Ofori K, Hernandez-Quijada K, Kim JK, Chung JH, Moore M, Moon SH, Seo JB, Kim YH. A Novel NOX Inhibitor Treatment Attenuates Parkinson's Disease-Related Pathology in Mouse Models. Int J Mol Sci 2022; 23:4262. [PMID: 35457082 PMCID: PMC9030373 DOI: 10.3390/ijms23084262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative motor disorder without an available therapeutic to halt the formation of Lewy bodies for preventing dopaminergic neuronal loss in the nigrostriatal pathway. Since oxidative-stress-mediated damage has been commonly reported as one of the main pathological mechanisms in PD, we assessed the efficacy of a novel NOX inhibitor from AptaBio Therapeutics (C-6) in dopaminergic cells and PD mouse models. The compound reduced the cytotoxicity and enhanced the cell viability at various concentrations against MPP+ and α-synuclein preformed fibrils (PFFs). Further, the levels of ROS and protein aggregation were significantly reduced at the optimal concentration (1 µM). Using two different mouse models, we gavaged C-6 at two different doses to the PD sign-displaying transgenic mice for 2 weeks and stereotaxically PFF-injected mice for 5 weeks. Our results demonstrated that both C-6-treated mouse models showed alleviated motor deficits in pole test, hindlimb clasping, crossbeam, rotarod, grooming, and nesting analyses. We also confirmed that the compound treatment reduced the levels of protein aggregation, along with phosphorylated-α-synuclein, in the striatum and ventral midbrain and further dopaminergic neuronal loss. Taken together, our results strongly suggest that NOX inhibition can be a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Anurupa A. Ghosh
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Dinesh Kumar Verma
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Gabriela Cabrera
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Kwadwo Ofori
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Karina Hernandez-Quijada
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Jae-Kwan Kim
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Joo Hee Chung
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Michael Moore
- Imaging Core, Delaware State University, Dover, DE 19901, USA;
| | - Sung Hwan Moon
- AptaBio Therapeutics Inc., 504 Tower, Heungdeok IT Valley, Heungdeok 1-ro 13, Gyeonggi-do, Yongin-si 16954, Korea;
| | - Jong Bok Seo
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Yong-Hwan Kim
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| |
Collapse
|
39
|
De Giorgi F, Abdul-Shukkoor MB, Kashyrina M, Largitte LA, De Nuccio F, Kauffmann B, Lends A, Laferrière F, Bonhommeau S, Lofrumento DD, Bousset L, Bezard E, Buffeteau T, Loquet A, Ichas F. Neurons with Cat's Eyes: A Synthetic Strain of α-Synuclein Fibrils Seeding Neuronal Intranuclear Inclusions. Biomolecules 2022; 12:436. [PMID: 35327628 PMCID: PMC8946814 DOI: 10.3390/biom12030436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
The distinct neuropathological features of the different α-Synucleinopathies, as well as the diversity of the α-Synuclein (α-Syn) intracellular inclusion bodies observed in post mortem brain sections, are thought to reflect the strain diversity characterizing invasive α-Syn amyloids. However, this "one strain, one disease" view is still hypothetical, and to date, a possible disease-specific contribution of non-amyloid factors has not been ruled out. In Multiple System Atrophy (MSA), the buildup of α-Syn inclusions in oligodendrocytes seems to result from the terminal storage of α-Syn amyloid aggregates first pre-assembled in neurons. This assembly occurs at the level of neuronal cytoplasmic inclusions, and even earlier, within neuronal intranuclear inclusions (NIIs). Intriguingly, α-Syn NIIs are never observed in α-Synucleinopathies other than MSA, suggesting that these inclusions originate (i) from the unique molecular properties of the α-Syn fibril strains encountered in this disease, or alternatively, (ii) from other factors specifically dysregulated in MSA and driving the intranuclear fibrillization of α-Syn. We report the isolation and structural characterization of a synthetic human α-Syn fibril strain uniquely capable of seeding α-Syn fibrillization inside the nuclear compartment. In primary mouse cortical neurons, this strain provokes the buildup of NIIs with a remarkable morphology reminiscent of cat's eye marbles (see video abstract). These α-Syn inclusions form giant patterns made of one, two, or three lentiform beams that span the whole intranuclear volume, pushing apart the chromatin. The input fibrils are no longer detectable inside the NIIs, where they become dominated by the aggregation of endogenous α-Syn. In addition to its phosphorylation at S129, α-Syn forming the NIIs acquires an epitope antibody reactivity profile that indicates its organization into fibrils, and is associated with the classical markers of α-Syn pathology p62 and ubiquitin. NIIs are also observed in vivo after intracerebral injection of the fibril strain in mice. Our data thus show that the ability to seed NIIs is a strain property that is integrally encoded in the fibril supramolecular architecture. Upstream alterations of cellular mechanisms are not required. In contrast to the lentiform TDP-43 NIIs, which are observed in certain frontotemporal dementias and which are conditional upon GRN or VCP mutations, our data support the hypothesis that the presence of α-Syn NIIs in MSA is instead purely amyloid-strain-dependent.
Collapse
Affiliation(s)
- Francesca De Giorgi
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, 33076 Bordeaux, France; (L.-A.L.); (F.L.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Muhammed Bilal Abdul-Shukkoor
- Institut de Chimie et de Biologie des Membranes et des Nano-objets, CNRS, UMR 5248, Université de Bordeaux, 33600 Pessac, France; (M.B.A.-S.); (A.L.); (A.L.)
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 33600 Pessac, France
| | - Marianna Kashyrina
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy; (M.K.); (F.D.N.); (D.D.L.)
| | - Leslie-Ann Largitte
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, 33076 Bordeaux, France; (L.-A.L.); (F.L.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francesco De Nuccio
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy; (M.K.); (F.D.N.); (D.D.L.)
| | - Brice Kauffmann
- Institut Européen de Chimie et Biologie, CNRS, Université de Bordeaux, INSERM, UMS3033/US001, 33600 Pessac, France;
| | - Alons Lends
- Institut de Chimie et de Biologie des Membranes et des Nano-objets, CNRS, UMR 5248, Université de Bordeaux, 33600 Pessac, France; (M.B.A.-S.); (A.L.); (A.L.)
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 33600 Pessac, France
| | - Florent Laferrière
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, 33076 Bordeaux, France; (L.-A.L.); (F.L.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Sébastien Bonhommeau
- Institut des Sciences Moléculaires, CNRS, UMR 5255, Université de Bordeaux, 33400 Talence, France; (S.B.); (T.B.)
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy; (M.K.); (F.D.N.); (D.D.L.)
| | - Luc Bousset
- Laboratory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, 92265 Fontenay-aux-Roses, France;
| | - Erwan Bezard
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, 33076 Bordeaux, France; (L.-A.L.); (F.L.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Thierry Buffeteau
- Institut des Sciences Moléculaires, CNRS, UMR 5255, Université de Bordeaux, 33400 Talence, France; (S.B.); (T.B.)
| | - Antoine Loquet
- Institut de Chimie et de Biologie des Membranes et des Nano-objets, CNRS, UMR 5248, Université de Bordeaux, 33600 Pessac, France; (M.B.A.-S.); (A.L.); (A.L.)
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 33600 Pessac, France
| | - François Ichas
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, 33076 Bordeaux, France; (L.-A.L.); (F.L.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| |
Collapse
|
40
|
Chen R, Gu X, Wang X. α-Synuclein in Parkinson's disease and advances in detection. Clin Chim Acta 2022; 529:76-86. [PMID: 35176268 DOI: 10.1016/j.cca.2022.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a threatening neurodegenerative disorder that seriously affects patients' life quality. Substantial evidence links the overexpression and abnormal aggregation of alpha-synuclein (α-Syn) to PD. α-Syn has been identified as a characteristic biomarker of PD, which indicates its great value of diagnosis and designing effective therapeutic strategy. This article systematically summarizes the pathogenic process of α-Syn based on recent researches, outlines and compares commonly used analysis and detection technologies of α-Syn. Specifically, the detection of α-Syn by new electrochemical, photochemical, and crystal biosensors is mainly examined. Furthermore, the speculation of future study orientation is discussed, which provides reference for the further research and application of α-Syn as biomarker.
Collapse
Affiliation(s)
- Rong Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xuan Gu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xiaoying Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
41
|
Lai TT, Kim YJ, Ma HI, Kim YE. Evidence of Inflammation in Parkinson’s Disease and Its Contribution to Synucleinopathy. J Mov Disord 2022; 15:1-14. [PMID: 35124957 PMCID: PMC8820875 DOI: 10.14802/jmd.21078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Accumulation of alpha-synuclein (αSyn) protein in neurons is a renowned pathological hallmark of Parkinson’s disease (PD). In addition, accumulating evidence indicates that activated inflammatory responses are involved in the pathogenesis of PD. Thus, achieving a better understanding of the interaction between inflammation and synucleinopathy in relation to the PD process will facilitate the development of promising disease-modifying therapies. In this review, the evidence of inflammation in PD is discussed, and human, animal, and laboratory studies relevant to the relationship between inflammation and αSyn are explored as well as new therapeutic targets associated with this relationship.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
- Hallym Neurological Institute, Hallym University College of Medicine, Anyang, Korea
| | - Yun Joong Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hyeo-il Ma
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
- Hallym Neurological Institute, Hallym University College of Medicine, Anyang, Korea
| | - Young Eun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
- Hallym Neurological Institute, Hallym University College of Medicine, Anyang, Korea
- Corresponding author: Young Eun Kim, MD Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, 22 Gwanpyeong-ro 170beon-gil, Dongangu, Anyang 14068, Korea / Tel: +82-31-380-3740 / E-mail:
| |
Collapse
|
42
|
Awa S, Suzuki G, Masuda-Suzukake M, Nonaka T, Saito M, Hasegawa M. Phosphorylation of endogenous α-synuclein induced by extracellular seeds initiates at the pre-synaptic region and spreads to the cell body. Sci Rep 2022; 12:1163. [PMID: 35064139 PMCID: PMC8782830 DOI: 10.1038/s41598-022-04780-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of phosphorylated α-synuclein aggregates has been implicated in several diseases, such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB), and is thought to spread in a prion-like manner. Elucidating the mechanisms of prion-like transmission of α-synuclein is important for the development of therapies for these diseases, but little is known about the details. Here, we injected α-synuclein fibrils into the brains of wild-type mice and examined the early phase of the induction of phosphorylated α-synuclein accumulation. We found that phosphorylated α-synuclein appeared within a few days after the intracerebral injection. It was observed initially in presynaptic regions and subsequently extended its localization to axons and cell bodies. These results suggest that extracellular α-synuclein fibrils are taken up into the presynaptic region and seed-dependently convert the endogenous normal α-synuclein that is abundant there to an abnormal phosphorylated form, which is then transported through the axon to the cell body.
Collapse
Affiliation(s)
- Shiori Awa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Genjiro Suzuki
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Masami Masuda-Suzukake
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Nonaka
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Minoru Saito
- Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan.,Department of Correlative Study in Physics and Chemistry, Graduate School of Integrated Basic Sciences, Nihon University, Tokyo, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
43
|
Vasili E, Dominguez-Meijide A, Flores-León M, Al-Azzani M, Kanellidi A, Melki R, Stefanis L, Outeiro TF. Endogenous Levels of Alpha-Synuclein Modulate Seeding and Aggregation in Cultured Cells. Mol Neurobiol 2022; 59:1273-1284. [PMID: 34984585 PMCID: PMC8857012 DOI: 10.1007/s12035-021-02713-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 11/30/2022]
Abstract
Parkinson’s disease is a progressive neurodegenerative disorder characterized by the accumulation of misfolded alpha-synuclein in intraneuronal inclusions known as Lewy bodies and Lewy neurites. Multiple studies strongly implicate the levels of alpha-synuclein as a major risk factor for the onset and progression of Parkinson’s disease. Alpha-synuclein pathology spreads progressively throughout interconnected brain regions but the precise molecular mechanisms underlying the seeding of alpha-synuclein aggregation are still unclear. Here, using stable cell lines expressing alpha-synuclein, we examined the correlation between endogenous alpha-synuclein levels and the seeding propensity by exogenous alpha-synuclein preformed fibrils. We applied biochemical approaches and imaging methods in stable cell lines expressing alpha-synuclein and in primary neurons to determine the impact of alpha-synuclein levels on seeding and aggregation. Our results indicate that the levels of alpha-synuclein define the pattern and severity of aggregation and the extent of p-alpha-synuclein deposition, likely explaining the selective vulnerability of different cell types in synucleinopathies. The elucidation of the cellular processes involved in the pathological aggregation of alpha-synuclein will enable the identification of novel targets and the development of therapeutic strategies for Parkinson’s disease and other synucleinopathies.
Collapse
Affiliation(s)
- Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073, Goettingen, Germany
| | - Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073, Goettingen, Germany
- Laboratory of Neuroanatomy and Experimental Neurology, Department. of Morphological Sciences, CIMUS, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Flores-León
- Departamento de Medicina Genómica Y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Mohammed Al-Azzani
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073, Goettingen, Germany
| | - Angeliki Kanellidi
- Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA, and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses, France
| | - Leonidas Stefanis
- Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073, Goettingen, Germany.
- Max Planck Institute for Experimental Medicine, Goettingen, Germany.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
- Scientific Employee With a Honorary Contract at Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.
| |
Collapse
|
44
|
Kang S, Piao Y, Kang YC, Lim S, Pak YK. DA-9805 protects dopaminergic neurons from endoplasmic reticulum stress and inflammation. Biomed Pharmacother 2022; 145:112389. [PMID: 34775235 DOI: 10.1016/j.biopha.2021.112389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 01/05/2023] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease with damages to mitochondria and endoplasmic reticulum (ER), followed by neuroinflammation. We previously reported that a triple herbal extract DA-9805 in experimental PD toxin-models had neuroprotective effects by alleviating mitochondrial damage and oxidative stress. In the present study, we investigated whether DA-9805 could suppress ER stress and neuroinflammation in vitro and/or in vivo. Pre-treatment with DA-9805 (1 μg/ml) attenuated upregulation of glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and cleaved caspase-3 in SH-SY5Y neuroblastoma cells treated with thapsigargin (1 µg/ml) or tunicamycin (2 µg/ml). In addition, DA-9805 prevented the production of IL-1β, IL-6, TNF-α and nitric oxide through inhibition of NF-κB activation in BV2 microglial cells stimulated with lipopolysaccharides (LPS). Intraperitoneal injection of LPS (10 mg/kg) into mice can induce acute neuroinflammation and dopaminergic neuronal cell death. Oral administration of DA-9805 (10 or 30 mg/kg/day for 3 days before LPS injection) prevented loss of dopaminergic neurons and activation of microglia and astrocytes in the substantia nigra in LPS-injected mouse models. Taken together, these results indicate that DA-9805 can effectively prevent ER stress and neuroinflammation, suggesting that DA-9805 is a multitargeting and disease-modifying therapeutic candidate for PD.
Collapse
Affiliation(s)
- Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Ying Piao
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Young Cheol Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Suyeol Lim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, South Korea; Department of Physiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea.
| |
Collapse
|
45
|
Stress-inducible phosphoprotein 1 (HOP/STI1/STIP1) regulates the accumulation and toxicity of α-synuclein in vivo. Acta Neuropathol 2022; 144:881-910. [PMID: 36121476 PMCID: PMC9547791 DOI: 10.1007/s00401-022-02491-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/26/2023]
Abstract
The predominantly pre-synaptic intrinsically disordered protein α-synuclein is prone to misfolding and aggregation in synucleinopathies, such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). Molecular chaperones play important roles in protein misfolding diseases and members of the chaperone machinery are often deposited in Lewy bodies. Here, we show that the Hsp90 co-chaperone STI1 co-immunoprecipitated α-synuclein, and co-deposited with Hsp90 and Hsp70 in insoluble protein fractions in two mouse models of α-synuclein misfolding. STI1 and Hsp90 also co-localized extensively with filamentous S129 phosphorylated α-synuclein in ubiquitin-positive inclusions. In PD human brains, STI1 transcripts were increased, and in neurologically healthy brains, STI1 and α-synuclein transcripts correlated. Nuclear Magnetic Resonance (NMR) analyses revealed direct interaction of α-synuclein with STI1 and indicated that the STI1 TPR2A, but not TPR1 or TPR2B domains, interacted with the C-terminal domain of α-synuclein. In vitro, the STI1 TPR2A domain facilitated S129 phosphorylation by Polo-like kinase 3. Moreover, mice over-expressing STI1 and Hsp90ß presented elevated α-synuclein S129 phosphorylation accompanied by inclusions when injected with α-synuclein pre-formed fibrils. In contrast, reduced STI1 function decreased protein inclusion formation, S129 α-synuclein phosphorylation, while mitigating motor and cognitive deficits as well as mesoscopic brain atrophy in α-synuclein-over-expressing mice. Our findings reveal a vicious cycle in which STI1 facilitates the generation and accumulation of toxic α-synuclein conformers, while α-synuclein-induced proteostatic stress increased insoluble STI1 and Hsp90.
Collapse
|
46
|
Costas C, Faro LR. Do Naturally Occurring Antioxidants Protect Against Neurodegeneration of the Dopaminergic System? A Systematic Revision in Animal Models of Parkinson's Disease. Curr Neuropharmacol 2022; 20:432-459. [PMID: 33882808 PMCID: PMC9413795 DOI: 10.2174/1570159x19666210421092725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by a significant decrease in dopamine levels, caused by progressive degeneration of the dopaminergic neurons in the nigrostriatal pathway. Multiple mechanisms have been implicated in its pathogenesis, including oxidative stress, neuroinflammation, protein aggregation, mitochondrial dysfunction, insufficient support for neurotrophic factors and cell apoptosis. The absence of treatments capable of slowing or stopping the progression of PD has increased the interest in the natural antioxidant substances present in the diet, since they have multiple beneficial properties and it is possible that they can influence the mechanisms responsible for the dysfunction and death of dopaminergic neurons. Thus, the purpose of this systematic review is to analyze the results obtained in a set of studies carried out in the last years, which describe the neuroprotective, antioxidant and regenerative functions of some naturally occurring antioxidants in experimental models of PD. The results show that the exogenous no enzymatic antioxidants can significantly modify the biochemical and behavioral mechanisms that contribute to the pathophysiology of Parkinsonism in experimental animals. Therefore, it is possible that they may contribute to effective neuroprotection by providing a significant improvement in neuropathological markers. In conclusion, the results of this review suggest that exogenous antioxidants can be promising therapeutic candidates for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Carmen Costas
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| | - Lilian R.F. Faro
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| |
Collapse
|
47
|
Zhang Q, Abdelmotilib H, Larson T, Keomanivong C, Conlon M, Aldridge GM, Narayanan NS. Cortical alpha-synuclein preformed fibrils do not affect interval timing in mice. Neurosci Lett 2021; 765:136273. [PMID: 34601038 DOI: 10.1016/j.neulet.2021.136273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
One hallmark feature of Parkinson's disease (PD) is Lewy body pathology associated with misfolded alpha-synuclein. Previous studies have shown that striatal injection of alpha-synuclein preformed fibrils (PFF) can induce misfolding and aggregation of native alpha-synuclein in a prion-like manner, leading to cell death and motor dysfunction in mouse models. Here, we tested whether alpha-synuclein PFFs injected into the medial prefrontal cortex results in deficits in interval timing, a cognitive task which is disrupted in human PD patients and in rodent models of PD. We injected PFF or monomers of human alpha-synuclein into the medial prefrontal cortex of mice pre-injected with adeno-associated virus (AAV) coding for overexpression of human alpha-synuclein or control protein. Despite notable medial prefrontal cortical synucleinopathy, we did not observe consistent deficits in fixed-interval timing. These results suggest that cortical alpha-synuclein does not reliably disrupt fixed-interval timing.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States.
| | - Hisham Abdelmotilib
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Travis Larson
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Cameron Keomanivong
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Mackenzie Conlon
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - Georgina M Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | | |
Collapse
|
48
|
Dauer Née Joppe K, Tatenhorst L, Caldi Gomes L, Zhang S, Parvaz M, Carboni E, Roser AE, El DeBakey H, Bähr M, Vogel-Mikuš K, Wang Ip C, Becker S, Zweckstetter M, Lingor P. Brain iron enrichment attenuates α-synuclein spreading after injection of preformed fibrils. J Neurochem 2021; 159:554-573. [PMID: 34176164 DOI: 10.1111/jnc.15461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
Regional iron accumulation and α-synuclein (α-syn) spreading pathology within the central nervous system are common pathological findings in Parkinson's disease (PD). Whereas iron is known to bind to α-syn, facilitating its aggregation and regulating α-syn expression, it remains unclear if and how iron also modulates α-syn spreading. To elucidate the influence of iron on the propagation of α-syn pathology, we investigated α-syn spreading after stereotactic injection of α-syn preformed fibrils (PFFs) into the striatum of mouse brains after neonatal brain iron enrichment. C57Bl/6J mouse pups received oral gavage with 60, 120, or 240 mg/kg carbonyl iron or vehicle between postnatal days 10 and 17. At 12 weeks of age, intrastriatal injections of 5-µg PFFs were performed to induce seeding of α-syn aggregates. At 90 days post-injection, PFFs-injected mice displayed long-term memory deficits, without affection of motor behavior. Interestingly, quantification of α-syn phosphorylated at S129 showed reduced α-syn pathology and attenuated spreading to connectome-specific brain regions after brain iron enrichment. Furthermore, PFFs injection caused intrastriatal microglia accumulation, which was alleviated by iron in a dose-dependent way. In primary cortical neurons in a microfluidic chamber model in vitro, iron application did not alter trans-synaptic α-syn propagation, possibly indicating an involvement of non-neuronal cells in this process. Our study suggests that α-syn PFFs may induce cognitive deficits in mice independent of iron. However, a redistribution of α-syn aggregate pathology and reduction of striatal microglia accumulation in the mouse brain may be mediated via iron-induced alterations of the brain connectome.
Collapse
Affiliation(s)
- Karina Dauer Née Joppe
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lucas Caldi Gomes
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Shuyu Zhang
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Mojan Parvaz
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Eleonora Carboni
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Anna-Elisa Roser
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Hazem El DeBakey
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Goettingen, Goettingen, Germany
| | - Katarina Vogel-Mikuš
- Biotechnical faculty, University of Ljubljana, Ljubljana, Slovenia
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Stefan Becker
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Markus Zweckstetter
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Research group Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
49
|
Lai TT, Kim YJ, Nguyen PT, Koh YH, Nguyen TT, Ma HI, Kim YE. Temporal Evolution of Inflammation and Neurodegeneration With Alpha-Synuclein Propagation in Parkinson's Disease Mouse Model. Front Integr Neurosci 2021; 15:715190. [PMID: 34675786 PMCID: PMC8523784 DOI: 10.3389/fnint.2021.715190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
According to a few studies, α-synuclein (αSyn) propagation has been suggested to play a key role in the pathomechanism of Parkinson's disease (PD), but neurodegeneration and the involvement of inflammation in its pathologic progression are not well understood with regard to temporal relationship. In this study, with the help of the PD mouse model injected with intrastriatal αSyn preformed fibril (PFF), the temporal evolution of αSyn propagation, inflammation, and neurodegeneration was explored in the perspective of the striatum and the whole brain. In the PFF-injected striatum, inflammatory response cells, including microglia and astrocytes, were activated at the earliest stage and reduced with time, and the phosphorylated form of αSyn accumulation increased behind it. Afterward, the degeneration of striatal dopaminergic neurons became significant with the conspicuity of behavioral phenotype. Similar patterns of forefront eruption of inflammation and then followed by αSyn propagation were noted in the opposite striatum, which were not injured by PFF injection. In analyzing the whole brain, inflammatory responses were activated at the earliest stage, and the soluble αSyn expression increased concurrently. The inflammatory response decreased afterward, and the accumulation of the insoluble form of αSyn increased behind it. Our results suggested that the inflammatory response may precede the accumulation of the pathologic form of αSyn; thereafter, the neurodegeneration and motor dysfunction followed αSyn proliferation in the PD mouse model. From this model, recognizing the temporal relationship between inflammation, αSyn propagation, and neurodegeneration may be helpful in establishing the PD animal model and monitoring the effect of interventional therapy.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| | - Yun Joong Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
| | - Phuong Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Young Ho Koh
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Tinh Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Hyeo-Il Ma
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| | - Young Eun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| |
Collapse
|
50
|
Malfertheiner K, Stefanova N, Heras-Garvin A. The Concept of α-Synuclein Strains and How Different Conformations May Explain Distinct Neurodegenerative Disorders. Front Neurol 2021; 12:737195. [PMID: 34675870 PMCID: PMC8523670 DOI: 10.3389/fneur.2021.737195] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
In the past few years, an increasing amount of studies primarily based on experimental models have investigated the existence of distinct α-synuclein strains and their different pathological effects. This novel concept could shed light on the heterogeneous nature of α-synucleinopathies, a group of disorders that includes Parkinson's disease, dementia with Lewy bodies and multiple system atrophy, which share as their key-molecular hallmark the abnormal aggregation of α-synuclein, a process that seems pivotal in disease pathogenesis according to experimental observations. However, the etiology of α-synucleinopathies and the initial events leading to the formation of α-synuclein aggregates remains elusive. Hence, the hypothesis that structurally distinct fibrillary assemblies of α-synuclein could have a causative role in the different disease phenotypes and explain, at least to some extent, their specific neurodegenerative, disease progression, and clinical presentation patterns is very appealing. Moreover, the presence of different α-synuclein strains might represent a potential biomarker for the diagnosis of these neurodegenerative disorders. In this regard, the recent use of super resolution techniques and protein aggregation assays has offered the possibility, on the one hand, to elucidate the conformation of α-synuclein pathogenic strains and, on the other hand, to cyclically amplify to detectable levels low amounts of α-synuclein strains in blood, cerebrospinal fluid and peripheral tissue from patients. Thus, the inclusion of these techniques could facilitate the differentiation between α-synucleinopathies, even at early stages, which is crucial for successful therapeutic intervention. This mini-review summarizes the current knowledge on α-synuclein strains and discusses its possible applications and potential benefits.
Collapse
Affiliation(s)
- Katja Malfertheiner
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonio Heras-Garvin
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|