1
|
Park H, Kam TI, Dawson VL, Dawson TM. α-Synuclein pathology as a target in neurodegenerative diseases. Nat Rev Neurol 2024:10.1038/s41582-024-01043-w. [PMID: 39609631 DOI: 10.1038/s41582-024-01043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
α-Synuclein misfolds into pathological forms that lead to various neurodegenerative diseases known collectively as α-synucleinopathies. In this Review, we provide a comprehensive overview of pivotal advances in α-synuclein research. We examine structural features and physiological functions of α-synuclein and summarize current insights into key post-translational modifications, such as nitration, phosphorylation, ubiquitination, sumoylation and truncation, considering their contributions to neurodegeneration. We also highlight the existence of disease-specific α-synuclein strains and their mechanisms of pathological spread, and discuss seed amplification assays and PET tracers as emerging diagnostic tools for detecting pathological α-synuclein in clinical settings. We also discuss α-synuclein aggregation and clearance mechanisms, and review cell-autonomous and non-cell-autonomous processes that contribute to neuronal death, including the roles of adaptive and innate immunity in α-synuclein-driven neurodegeneration. Finally, we highlight promising therapeutic approaches that target pathological α-synuclein and provide insights into emerging areas of research.
Collapse
Affiliation(s)
- Hyejin Park
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Chahardehi AM, Hosseini Y, Mahdavi SM, Naseh I. The Zebrafish Model as a New Discovery Path for Medicinal Plants in the Treatment of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:306-314. [PMID: 36999188 DOI: 10.2174/1871527322666230330111712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 04/01/2023]
Abstract
Parkinson's disease (PD) is one of the most frequent degenerative central nervous system disorders affecting older adults. Dopaminergic neuron failure in the substantia nigra is a pathological sign connected with the motor shortfall of PD. Due to their low teratogenic and adverse effect potential, medicinal herbs have emerged as a promising therapy option for preventing and curing PD and other neurodegenerative disorders. However, the mechanism through which natural compounds provide neuroprotection against PD remains unknown. While testing compounds in vertebrates such as mice is prohibitively expensive and time-consuming, zebrafish (Danio rerio) may offer an appealing alternative because they are vertebrates and share many of the same characteristics as humans. Zebrafish are commonly used as animal models for studying many human diseases, and their molecular history and bioimaging properties are appropriate for the study of PD. However, a literature review indicated that only six plants, including Alpinia oxyhylla, Bacopa monnieri, Canavalia gladiate, Centella asiatica, Paeonia suffruticosa, and Stachytarpheta indica had been investigated as potential PD treatments using the zebrafish model. Only C. asiatica and B. monnieri were found to have potential anti-PD activity. In addition to reviewing the current state of research in this field, these plants' putative mechanisms of action against PD are explored, and accessible assays for investigation are made.
Collapse
Affiliation(s)
| | - Yasaman Hosseini
- Cognitive Neuroscience Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Mahdavi
- Department of Bioscience and Biotechnology, Malek Ashtar University of Technology (MUT), Tehran, Iran
| | - Iman Naseh
- Cognitive Neuroscience Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Peggion C, Barazzuol L, Poggio E, Calì T, Brini M. Ca 2+ signalling: A common language for organelles crosstalk in Parkinson's disease. Cell Calcium 2023; 115:102783. [PMID: 37597300 DOI: 10.1016/j.ceca.2023.102783] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/21/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease caused by multifactorial pathogenic mechanisms. Familial PD is linked with genetic mutations in genes whose products are either associated with mitochondrial function or endo-lysosomal pathways. Of note, mitochondria are essential to sustain high energy demanding synaptic activity of neurons and alterations in mitochondrial Ca2+ signaling have been proposed as causal events for neurodegenerative process, although the mechanisms responsible for the selective loss of specific neuronal populations in the different neurodegenerative diseases is still not clear. Here, we specifically discuss the importance of a correct mitochondrial communication with the other organelles occurring at regions where their membranes become in close contact. We discuss the nature and the role of contact sites that mitochondria establish with ER, lysosomes, and peroxisomes, and how PD related proteins participate in the regulation/dysregulation of the tethering complexes. Unravelling molecular details of mitochondria tethering could contribute to identify specific therapeutic targets and develop new strategies to counteract the progression of the disease.
Collapse
Affiliation(s)
| | | | - Elena Poggio
- Department of Biology (DIBIO), University of Padova, Italy
| | - Tito Calì
- Department of Biomedical Sciences (DSB), University of Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Italy; Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
| | - Marisa Brini
- Department of Biology (DIBIO), University of Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Italy.
| |
Collapse
|
4
|
Mackie PM, Koshy J, Bhogade M, Hammoor T, Hachmeister W, Lloyd GM, Paterno G, Bolen M, Tansey MG, Giasson BI, Khoshbouei H. Complement C1q-dependent engulfment of alpha-synuclein induces ENS-resident macrophage exhaustion and accelerates Parkinson's-like gut pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563832. [PMID: 37961460 PMCID: PMC10634831 DOI: 10.1101/2023.10.24.563832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Deposition of misfolded α-synuclein (αsyn) in the enteric nervous system (ENS) is found in multiple neurodegenerative diseases. It is hypothesized that ENS synucleinopathy contributes to both the pathogenesis and non-motor morbidity in Parkinson's Disease (PD), but the cellular and molecular mechanisms that shape enteric histopathology and dysfunction are poorly understood. Here, we demonstrate that ENS-resident macrophages, which play a critical role in maintaining ENS homeostasis, initially respond to enteric neuronal αsyn pathology by upregulating machinery for complement-mediated engulfment. Pharmacologic depletion of ENS-macrophages or genetic deletion of C1q enhanced enteric neuropathology. Conversely, C1q deletion ameliorated gut dysfunction, indicating that complement partially mediates αsyn-induced gut dysfunction. Internalization of αsyn led to increased endo-lysosomal stress that resulted in macrophage exhaustion and temporally correlated with the progression of ENS pathology. These novel findings highlight the importance of enteric neuron-macrophage interactions in removing toxic protein aggregates that putatively shape the earliest stages of PD in the periphery.
Collapse
Affiliation(s)
- P M Mackie
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - J Koshy
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M Bhogade
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - T Hammoor
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - W Hachmeister
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - G M Lloyd
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - G Paterno
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M Bolen
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M G Tansey
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, Florida, 32610
| | - B I Giasson
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - H Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| |
Collapse
|
5
|
Sant'Anna R, Robbs BK, de Freitas JA, Dos Santos PP, König A, Outeiro TF, Foguel D. The alpha-synuclein oligomers activate nuclear factor of activated T-cell (NFAT) modulating synaptic homeostasis and apoptosis. Mol Med 2023; 29:111. [PMID: 37596531 PMCID: PMC10439599 DOI: 10.1186/s10020-023-00704-8] [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: 03/31/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Soluble oligomeric forms of alpha-synuclein (aSyn-O) are believed to be one of the main toxic species in Parkinson's disease (PD) leading to degeneration. aSyn-O can induce Ca2+ influx, over activating downstream pathways leading to PD phenotype. Calcineurin (CN), a phosphatase regulated by Ca2+ levels, activates NFAT transcription factors that are involved in the regulation of neuronal plasticity, growth, and survival. METHODS Here, using a combination of cell toxicity and gene regulation assays performed in the presence of classical inhibitors of the NFAT/CN pathway, we investigate NFAT's role in neuronal degeneration induced by aSyn-O. RESULTS aSyn-O are toxic to neurons leading to cell death, loss of neuron ramification and reduction of synaptic proteins which are reversed by CN inhibition with ciclosporin-A or VIVIT, a NFAT specific inhibitor. aSyn-O induce NFAT nuclear translocation and transactivation. We found that aSyn-O modulates the gene involved in the maintenance of synapses, synapsin 1 (Syn 1). Syn1 mRNA and protein and synaptic puncta are drastically reduced in cells treated with aSyn-O which are reversed by NFAT inhibition. CONCLUSIONS For the first time a direct role of NFAT in aSyn-O-induced toxicity and Syn1 gene regulation was demonstrated, enlarging our understanding of the pathways underpinnings synucleinopathies.
Collapse
Affiliation(s)
- Ricardo Sant'Anna
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Centro de Ciências da Saúde, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Bloco E sala 42, Rio de Janeiro, 21941-590, Brazil
| | - Bruno K Robbs
- Departamento de Ciência Básica, Instituto de Saúde de Nova Friburgo, Universidade Federal Fluminense, Nova Friburgo, RJ, 28625-650, Brazil
| | - Júlia Araújo de Freitas
- Centro de Ciências da Saúde, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Bloco E sala 42, Rio de Janeiro, 21941-590, Brazil
| | - Patrícia Pires Dos Santos
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany.
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.
| | - Debora Foguel
- Centro de Ciências da Saúde, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Bloco E sala 42, Rio de Janeiro, 21941-590, Brazil.
| |
Collapse
|
6
|
Kaya I, Schembri LS, Nilsson A, Shariatgorji R, Baijnath S, Zhang X, Bezard E, Svenningsson P, Odell LR, Andrén PE. On-Tissue Chemical Derivatization for Comprehensive Mapping of Brain Carboxyl and Aldehyde Metabolites by MALDI-MS Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:836-846. [PMID: 37052344 PMCID: PMC10161219 DOI: 10.1021/jasms.2c00336] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The visualization of small metabolites by MALDI mass spectrometry imaging in brain tissue sections is challenging due to low detection sensitivity and high background interference. We present an on-tissue chemical derivatization MALDI mass spectrometry imaging approach for the comprehensive mapping of carboxyls and aldehydes in brain tissue sections. In this approach, the AMPP (1-(4-(aminomethyl)phenyl)pyridin-1-ium chloride) derivatization reagent is used for the covalent charge-tagging of molecules containing carboxylic acid (in the presence of peptide coupling reagents) and aldehydes. This includes free fatty acids and the associated metabolites, fatty aldehydes, dipeptides, neurotoxic reactive aldehydes, amino acids, neurotransmitters and associated metabolites, as well as tricarboxylic acid cycle metabolites. We performed sensitive ultrahigh mass resolution MALDI-MS detection and imaging of various carboxyl- and aldehyde-containing endogenous metabolites simultaneously in rodent brain tissue sections. We verified the AMPP-derivatized metabolites by tandem MS for structural elucidation. This approach allowed us to image numerous aldehydes and carboxyls, including certain metabolites which had been undetectable in brain tissue sections. We also demonstrated the application of on-tissue derivatization to carboxyls and aldehydes in coronal brain tissue sections of a nonhuman primate Parkinson's disease model. Our methodology provides a powerful tool for the sensitive, simultaneous spatial molecular imaging of numerous aldehydes and carboxylic acids during pathological states, including neurodegeneration, in brain tissue.
Collapse
Affiliation(s)
- Ibrahim Kaya
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| | | | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| | - Sooraj Baijnath
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| | - Xiaoqun Zhang
- Section of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France
| | - Per Svenningsson
- Section of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Luke R Odell
- Department of Medicinal Chemistry, Uppsala University, SE-75123 Uppsala, Sweden
| | - Per E Andrén
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| |
Collapse
|
7
|
Liu Z, Huang Y, Wang X, Li JY, Zhang C, Yang Y, Zhang J. The cervical lymph node contributes to peripheral inflammation related to Parkinson's disease. J Neuroinflammation 2023; 20:93. [PMID: 37038192 PMCID: PMC10088204 DOI: 10.1186/s12974-023-02770-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/20/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Peripheral inflammation is an important feature of Parkinson's disease (PD). However, if and how CNS pathology is involved in the peripheral inflammation in PD remains to be fully investigated. Recently, the existence of meningeal lymphatics and its involvement in draining cerebral spinal fluid (CSF) to the cervical lymph node has been discovered. It is known that meningeal lymphatic dysfunction exists in idiopathic PD. The deep cervical lymph node (dCLN) substantially contributes to the drainage of the meningeal lymphatics. In addition, one of the lymphatics draining components, CSF, contains abundant α-synuclein (α-syn), a protein critically involved in PD pathogenesis and neuroinflammation. Thus, we began with exploring the possible structural and functional alterations of the dCLN in a PD mouse model (A53T mice) and investigated the role of pathological α-syn in peripheral inflammation and its potential underlying molecular mechanisms. METHODS In this study, the transgenic mice (prnp-SNCA*A53T) which specifically overexpressed A53T mutant α-syn in CNS were employed as the PD animal model. Immunofluorescent and Hematoxylin and eosin staining were used to evaluate structure of dCLN. Inflammation in dCLNs as well as in bone-marrow-derived macrophages (BMDMs) was assessed quantitatively by measuring the mRNA and protein levels of typical inflammatory cytokines (including IL-1β, IL-6 and TNF-α). Intra-cisterna magna injection, flow cytometric sorting and electrochemiluminescence immunoassays were applied to investigate the lymphatic drainage of α-syn from the CNS. RNA-seq and Western blot were used to explore how pathological α-syn mediated the inflammation in PD mice. RESULTS The results unequivocally revealed substantially enlarged dCLNs, along with slow lymphatic flow, and increased inflammation in the dCLNs of A53T mice. Oligomeric α-syn drained from CSF potently activated macrophages in the dCLN via endoplasmic reticulum (ER) stress. Notably, inhibition of ER stress effectively suppressed peripheral inflammation in PD mice. CONCLUSIONS Our findings indicate that lymph node enlargement is closely related to macrophage activation, induced by meningeal lymphatics draining oligomeric α-syn, and contributes to the peripheral inflammation in PD. In addition, ER stress is a potential therapeutic target to ameliorate PD pathogenesis.
Collapse
Affiliation(s)
- Zongran Liu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | - Yang Huang
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | - Xuejing Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Institute of Health Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Can Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Ying Yang
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China.
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China.
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
8
|
Peña-Díaz S, García-Pardo J, Ventura S. Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson's Disease. Pharmaceutics 2023; 15:839. [PMID: 36986700 PMCID: PMC10059018 DOI: 10.3390/pharmaceutics15030839] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Parkinson's disease, the second most common neurodegenerative disorder worldwide, is characterized by the accumulation of protein deposits in the dopaminergic neurons. These deposits are primarily composed of aggregated forms of α-Synuclein (α-Syn). Despite the extensive research on this disease, only symptomatic treatments are currently available. However, in recent years, several compounds, mainly of an aromatic character, targeting α-Syn self-assembly and amyloid formation have been identified. These compounds, discovered by different approaches, are chemically diverse and exhibit a plethora of mechanisms of action. This work aims to provide a historical overview of the physiopathology and molecular aspects associated with Parkinson's disease and the current trends in small compound development to target α-Syn aggregation. Although these molecules are still under development, they constitute an important step toward discovering effective anti-aggregational therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Javier García-Pardo
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
9
|
Brsikyan LA, Poluektova EA, Poluektov MG. The gut microbiome as a factor in the development of Parkinson's disease. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2023. [DOI: 10.14412/2074-2711-2023-1-90-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- L. A. Brsikyan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | - E. A. Poluektova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | - M. G. Poluektov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| |
Collapse
|
10
|
Neupane S, De Cecco E, Aguzzi A. The Hidden Cell-to-Cell Trail of α-Synuclein Aggregates. J Mol Biol 2022:167930. [PMID: 36566800 DOI: 10.1016/j.jmb.2022.167930] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
The progressive accumulation of insoluble aggregates of the presynaptic protein alpha-synuclein (α-Syn) is a hallmark of neurodegenerative disorders including Parkinson's disease (PD), Multiple System Atrophy, and Dementia with Lewy Bodies, commonly referred to as synucleinopathies. Despite considerable progress on the structural biology of these aggregates, the molecular mechanisms mediating their cell-to-cell transmission, propagation, and neurotoxicity remain only partially understood. Numerous studies have highlighted the stereotypical spatiotemporal spreading of pathological α-Syn aggregates across different tissues and anatomically connected brain regions over time. Experimental evidence from various cellular and animal models indicate that α-Syn transfer occurs in two defined steps: the release of pathogenic α-Syn species from infected cells, and their uptake via passive or active endocytic pathways. Once α-Syn aggregates have been internalized, little is known about what drives their toxicity or how they interact with the endogenous protein to promote its misfolding and subsequent aggregation. Similarly, unknown genetic factors modulate different cellular responses to the aggregation and accumulation of pathogenic α-Syn species. Here we discuss the current understanding of the molecular phenomena associated with the intercellular spreading of pathogenic α-Syn seeds and summarize the evidence supporting the transmission hypothesis. Understanding the molecular mechanisms involved in α-Syn aggregates transmission is essential to develop novel targeted therapeutics against PD and related synucleinopathies.
Collapse
Affiliation(s)
- Sandesh Neupane
- Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland. https://twitter.com/neuron_sandesh
| | - Elena De Cecco
- Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland.
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland.
| |
Collapse
|
11
|
Vestuto V, Di Sarno V, Musella S, Di Dona G, Moltedo O, Gomez-Monterrey IM, Bertamino A, Ostacolo C, Campiglia P, Ciaglia T. New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors? Int J Mol Sci 2022; 24:185. [PMID: 36613628 PMCID: PMC9820239 DOI: 10.3390/ijms24010185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic structure, playing multiple roles including calcium storage, protein synthesis and lipid metabolism. During cellular stress, variations in ER homeostasis and its functioning occur. This condition is referred as ER stress and generates a cascade of signaling events termed unfolded protein response (UPR), activated as adaptative response to mitigate the ER stress condition. In this regard, calcium levels play a pivotal role in ER homeostasis and therefore in cell fate regulation since calcium signaling is implicated in a plethora of physiological processes, but also in disease conditions such as neurodegeneration, cancer and metabolic disorders. A large body of emerging evidence highlighted the functional role of TRP channels and their ability to promote cell survival or death depending on endoplasmic reticulum stress resolution, making them an attractive target. Thus, in this review we focused on the TRP channels' correlation to UPR-mediated ER stress in disease pathogenesis, providing an overview of their implication in the activation of this cellular response.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Giorgio Di Dona
- Pineta Grande Hospital, Via Domiziana, km 30/00, 81030 Castel Volturno, CE, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | | | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- European Biomedical Research Institute of Salerno, Via S. De Renzi 50, 84125 Salerno, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| |
Collapse
|
12
|
Wilhelm E, Quoilin C, Derosiere G, Paço S, Jeanjean A, Duque J. Corticospinal Suppression Underlying Intact Movement Preparation Fades in Parkinson's Disease. Mov Disord 2022; 37:2396-2406. [PMID: 36121426 DOI: 10.1002/mds.29214] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In Parkinson's disease (PD), neurophysiological abnormalities within the primary motor cortex (M1) have been shown to contribute to bradykinesia, but exact modalities are still uncertain. We propose that such motor slowness could involve alterations in mechanisms underlying movement preparation, especially the suppression of corticospinal excitability-called "preparatory suppression"-which is considered to propel movement execution by increasing motor neural gain in healthy individuals. METHODS On two consecutive days, 29 PD patients (on and off medication) and 29 matched healthy controls (HCs) underwent transcranial magnetic stimulation over M1, eliciting motor-evoked potentials (MEPs) in targeted hand muscles, while they were either at rest or preparing a left- or right-hand response in an instructed-delay choice reaction time task. Preparatory suppression was assessed by expressing MEP amplitudes during movement preparation relative to rest. RESULTS Contrary to HCs, PD patients showed a lack of preparatory suppression when the side of the responding hand was analyzed, especially when the latter was the most affected one. This deficit, which did not depend on dopamine medication, increased with disease duration and also tended to correlate with motor impairment, as measured by the Movement Disorder Society Unified Parkinson's Disease Rating Scale, Part III (both total and bradykinesia scores). CONCLUSIONS Our novel findings indicate that preparatory suppression fades in PD, in parallel with worsening motor symptoms, including bradykinesia. Such results suggest that an alteration in this marker of intact movement preparation could indeed cause motor slowness and support its use in future studies on the relation between M1 alterations and motor impairment in PD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Emmanuelle Wilhelm
- CoActions Lab, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Department of Adult Neurology, Saint-Luc University Hospital, Brussels, Belgium
| | - Caroline Quoilin
- CoActions Lab, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Gerard Derosiere
- CoActions Lab, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Susana Paço
- NOVA IMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Anne Jeanjean
- Department of Adult Neurology, Saint-Luc University Hospital, Brussels, Belgium
| | - Julie Duque
- CoActions Lab, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
13
|
Better Outcomes with Intranigral versus Intrastriatal Cell Transplantation: Relevance for Parkinson’s Disease. Cells 2022; 11:cells11071191. [PMID: 35406755 PMCID: PMC8997951 DOI: 10.3390/cells11071191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Intrastriatal embryonic ventral mesencephalon grafts have been shown to integrate, survive, and reinnervate the host striatum in clinical settings and in animal models of Parkinson’s disease. However, this ectopic location does not restore the physiological loops of the nigrostriatal pathway and promotes only moderate behavioral benefits. Here, we performed a direct comparison of the potential benefits of intranigral versus intrastriatal grafts in animal models of Parkinson’s disease. We report that intranigral grafts promoted better survival of dopaminergic neurons and that only intranigral grafts induced recovery of fine motor skills and normalized cortico-striatal responses. The increase in the number of toxic activated glial cells in host tissue surrounding the intrastriatal graft, as well as within the graft, may be one of the causes of the increased cell death observed in the intrastriatal graft. Homotopic localization of the graft and the subsequent physiological cell rewiring of the basal ganglia may be a key factor in successful and beneficial cell transplantation procedures.
Collapse
|
14
|
Choong CJ, Mochizuki H. Neuropathology of α-synuclein in Parkinson's disease. Neuropathology 2022; 42:93-103. [PMID: 35362115 DOI: 10.1111/neup.12812] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 01/21/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive movement disability accompanied by non-motor symptoms. The neuropathology hallmark of PD is the loss of dopaminergic neurons predominantly in the substantia nigra pars compacta and the presence of intracellular inclusions termed Lewy bodies (LBs), which are mainly composed of α-synuclein (αSyn). Detailed staging based on the distribution and progression pattern of αSyn pathology in the postmortem brains of PD patients revealed correlation with the clinical phenotypes but not invariably. Cumulative evidence from cell and animal studies has implied that αSyn propagation contributes to the anatomical spread of αSyn pathology in the brain. Here, we recount the studies over the past two centuries on the anatomopathological foundations of PD documented. We also review studies on the structural analysis of αSyn and LBs, Braak staging of αSyn pathology, the cell-to-cell propagation of αSyn as well as αSyn fibril polymorphisms, which underlie the phenotypic differences in synucleinopathies.
Collapse
Affiliation(s)
- Chi-Jing Choong
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
15
|
Sano T, Kawazoe T, Shioya A, Mori-Yoshimura M, Oya Y, Maruo K, Nishino I, Hoshino M, Murayama S, Saito Y. Unique Lewy pathology in myotonic dystrophy type 1. Neuropathology 2022; 42:104-116. [PMID: 35199386 DOI: 10.1111/neup.12790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 01/25/2023]
Abstract
Lewy body-related α-synucleinopathy (Lewy pathology) has been reported in patients with myotonic dystrophy (DM) type 1 (DM1), but no detailed report has described the prevalence and extent of its occurrence. We studied consecutive full autopsy cases of DM1 at the National Center of Neurology and Psychiatry (NCNP) Brain Bank for intractable psychiatric and neurological disorders. Thirty-two cases, genetically determined to be DM1 (59.0 ± 8.7 years), obtained from the NCNP Brain Bank, were compared with control cases obtained from the Brain Bank for Aging Research (BBAR) in Japan. The investigated anatomical sites followed the Dementia with Lewy Bodies Consensus Guideline, expanding to the peripheral autonomic nervous system, temporal pole, and occipital cortex, in addition to the olfactory epithelium and spinal cord. Of the 32 patients, 11 (34.4%) had Lewy pathology, with a significantly higher prevalence than that in the control cases from the BBAR (20.1%). Lewy pathology detected in DM1 was widespread, but no macroscopic depigmentation of the substantia nigra was observed in any DM1 case; this was commensurate with the microscopic paucity of Lewy pathology in the substantia nigra and amygdala. Lewy pathology in DM1 does not appear to follow either Braak's ascending paradigm or the olfactory-amygdala extension. Lewy neurites and dots in DM1 were very sparse in the cerebral cortex and distinct from those observed in BBAR control cases. This study was the first demonstration of unique Lewy pathology in DM1 and may contribute to the understanding of the protein propagation hypothesis of Lewy pathology.
Collapse
Affiliation(s)
- Terunori Sano
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan.,NCNP Brain Physiology and Pathology, Cognitive and Behavioral Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoya Kawazoe
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ayako Shioya
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, Mito Kyodo General Hospital, Tsukuba University Hospital Mito Area Medical Education Center, Ibaraki, Japan
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yasushi Oya
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kazushi Maruo
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Ichizo Nishino
- Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mikio Hoshino
- NCNP Brain Physiology and Pathology, Cognitive and Behavioral Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neuropathology and Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan.,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Yuko Saito
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neuropathology and Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
16
|
Gouda NA, Elkamhawy A, Cho J. Emerging Therapeutic Strategies for Parkinson’s Disease and Future Prospects: A 2021 Update. Biomedicines 2022; 10:biomedicines10020371. [PMID: 35203580 PMCID: PMC8962417 DOI: 10.3390/biomedicines10020371] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder pathologically distinguished by degeneration of dopaminergic neurons in the substantia nigra pars compacta. Muscle rigidity, tremor, and bradykinesia are all clinical motor hallmarks of PD. Several pathways have been implicated in PD etiology, including mitochondrial dysfunction, impaired protein clearance, and neuroinflammation, but how these factors interact remains incompletely understood. Although many breakthroughs in PD therapy have been accomplished, there is currently no cure for PD, only trials to alleviate the related motor symptoms. To reduce or stop the clinical progression and mobility impairment, a disease-modifying approach that can directly target the etiology rather than offering symptomatic alleviation remains a major unmet clinical need in the management of PD. In this review, we briefly introduce current treatments and pathophysiology of PD. In addition, we address the novel innovative therapeutic targets for PD therapy, including α-synuclein, autophagy, neurodegeneration, neuroinflammation, and others. Several immunomodulatory approaches and stem cell research currently in clinical trials with PD patients are also discussed. Moreover, preclinical studies and clinical trials evaluating the efficacy of novel and repurposed therapeutic agents and their pragmatic applications with encouraging outcomes are summarized. Finally, molecular biomarkers under active investigation are presented as potentially valuable tools for early PD diagnosis.
Collapse
Affiliation(s)
- Noha A. Gouda
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
| | - Ahmed Elkamhawy
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Jungsook Cho
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Correspondence:
| |
Collapse
|
17
|
Spathopoulou A, Edenhofer F, Fellner L. Targeting α-Synuclein in Parkinson's Disease by Induced Pluripotent Stem Cell Models. Front Neurol 2022; 12:786835. [PMID: 35145469 PMCID: PMC8821105 DOI: 10.3389/fneur.2021.786835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder characterized by motor and non-motor symptoms. To date, no specific treatment to halt disease progression is available, only medication to alleviate symptoms can be prescribed. The main pathological hallmark of PD is the development of neuronal inclusions, positive for α-synuclein (α-syn), which are termed Lewy bodies (LBs) or Lewy neurites. However, the cause of the inclusion formation and the loss of neurons remain largely elusive. Various genetic determinants were reported to be involved in PD etiology, including SNCA, DJ-1, PRKN, PINK1, LRRK2, and GBA. Comprehensive insights into pathophysiology of PD critically depend on appropriate models. However, conventional model organisms fall short to faithfully recapitulate some features of this complex disease and as a matter-of-fact access to physiological tissue is limiting. The development of disease models replicating PD that are close to human physiology and dynamic enough to analyze the underlying molecular mechanisms of disease initiation and progression, as well as the generation of new treatment options, is an important and overdue step. Recently, the establishment of induced pluripotent stem cell (iPSC)-derived neural models, particularly from genetic PD-variants, developed into a promising strategy to investigate the molecular mechanisms regarding formation of inclusions and neurodegeneration. As these iPSC-derived neurons can be generated from accessible biopsied samples of PD patients, they carry pathological alterations and enable the possibility to analyze the differences compared to healthy neurons. This review focuses on iPSC models carrying genetic PD-variants of α-syn that will be especially helpful in elucidating the pathophysiological mechanisms of PD. Furthermore, we discuss how iPSC models can be instrumental in identifying cellular targets, potentially leading to the development of new therapeutic treatments. We will outline the enormous potential, but also discuss the limitations of iPSC-based α-syn models.
Collapse
|
18
|
Barker RA. The Origins of the Protein Spread in Parkinson's Disease. Mov Disord Clin Pract 2021; 8:1194-1197. [PMID: 34765685 DOI: 10.1002/mdc3.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/05/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Roger A Barker
- Wellcome-MRC Stem Cell institute and Department of Neurology, Addenbrooke's Hospital University of Cambridge Cambridge United Kingdom
| |
Collapse
|
19
|
Singh A, Maharana SK, Shukla R, Kesharwani P. Nanotherapeutics approaches for targeting alpha synuclien protein in the management of Parkinson disease. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.08.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
20
|
Scheiblich H, Dansokho C, Mercan D, Schmidt SV, Bousset L, Wischhof L, Eikens F, Odainic A, Spitzer J, Griep A, Schwartz S, Bano D, Latz E, Melki R, Heneka MT. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell 2021; 184:5089-5106.e21. [PMID: 34555357 PMCID: PMC8527836 DOI: 10.1016/j.cell.2021.09.007] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 06/05/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022]
Abstract
Microglia are the CNS resident immune cells that react to misfolded proteins through pattern recognition receptor ligation and activation of inflammatory pathways. Here, we studied how microglia handle and cope with α-synuclein (α-syn) fibrils and their clearance. We found that microglia exposed to α-syn establish a cellular network through the formation of F-actin-dependent intercellular connections, which transfer α-syn from overloaded microglia to neighboring naive microglia where the α-syn cargo got rapidly and effectively degraded. Lowering the α-syn burden attenuated the inflammatory profile of microglia and improved their survival. This degradation strategy was compromised in cells carrying the LRRK2 G2019S mutation. We confirmed the intercellular transfer of α-syn assemblies in microglia using organotypic slice cultures, 2-photon microscopy, and neuropathology of patients. Together, these data identify a mechanism by which microglia create an "on-demand" functional network in order to improve pathogenic α-syn clearance.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Cira Dansokho
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Dilek Mercan
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Luc Bousset
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-aux-Roses, France
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Frederik Eikens
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Alexandru Odainic
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Jasper Spitzer
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Stephanie Schwartz
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-aux-Roses, France
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605 Worcester, MA, USA.
| |
Collapse
|
21
|
Persons AL, Bradaric BD, Kelly LP, Kousik SM, Graves SM, Yamamoto BK, Napier TC. Gut and brain profiles that resemble pre-motor and early-stage Parkinson's disease in methamphetamine self-administering rats. Drug Alcohol Depend 2021; 225:108746. [PMID: 34098381 PMCID: PMC8483557 DOI: 10.1016/j.drugalcdep.2021.108746] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Methamphetamine is a potent psychomotor stimulant, and methamphetamine abusers are up to three times more likely to develop Parkinson's disease (PD) later in life. Prodromal PD may involve gut inflammation and the accumulation of toxic proteins that are transported from the enteric nervous system to the central nervous system to mediate, in part, the degeneration of dopaminergic projections. We hypothesized that self-administration of methamphetamine in rats produces a gut and brain profile that mirrors pre-motor and early-stage PD. METHODS Rats self-administered methamphetamine in daily 3 h sessions for two weeks. Motor function was assessed before self-administration, during self-administration and throughout the 56 days of forced abstinence. Assays for pathogenic markers (tyrosine hydroxylase, glial fibrillary acidic protein (GFAP), α-synuclein) were conducted on brain and gut tissue collected at one or 56 days after cessation of methamphetamine self-administration. RESULTS Motor deficits emerged by day 14 of forced abstinence and progressively worsened up to 56 days of forced abstinence. In the pre-motor stage, we observed increased immunoreactivity for GFAP and α-synuclein within the ganglia of the myenteric plexus in the distal colon. Increased α-synuclein was also observed in the substantia nigra pars compacta. At 56 days, GFAP and α-synuclein normalized in the gut, but the accumulation of nigral α-synuclein persisted, and the dorsolateral striatum exhibited a significant loss of tyrosine hydroxylase. CONCLUSION The pre-motor profile is consistent with gut inflammation and gut/brain α-synuclein accumulation associated with prodromal PD and the eventual development of the neurological disease.
Collapse
Affiliation(s)
- Amanda L. Persons
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612,Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL 60612,Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL 60612,Department of Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612
| | - Brinda D. Bradaric
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612,Department of Health Sciences, Rush University Medical Center, Chicago, IL 60612,Department of Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612
| | - Leo P. Kelly
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612
| | - Sharanya M. Kousik
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612,Department of Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612
| | - Steven M. Graves
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612,Department of Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612
| | - Bryan K. Yamamoto
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - T. Celeste Napier
- Department of Pharmacology, Rush University Medical Center, Chicago, IL 60612,Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL 60612,Department of Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
22
|
Zheng T, Zhang Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson's disease. Neurochem Int 2021; 148:105094. [PMID: 34097990 DOI: 10.1016/j.neuint.2021.105094] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and abnormal aggregates of α-synuclein protein called Lewy bodies. To date, there is no drug that can definitely slow down or stop the progression of this disease. The discovery of the cell-to-cell transmission of pathologic α-synuclein seeds offers the possibility to explore novel treatment strategies to prevent the spread of α-synuclein, with the purpose of slowing down the progression of PD in its tracks. Although recent studies have made tremendous progress in understanding how α-synuclein spreads throughout the brain, neuroinflammation seems to play a crucial role in the development of α-synuclein pathology in PD. The activation of microglia, one of the hallmarks of the neuroinflammatory process, is suggested to influence the neuron-to-neuron transmission of α-synuclein. This review summarizes how activated microglia facilitate this process, and focuses on the following mechanisms including the activation of microglia in PD, the reduced ability of activated microglia to clear α-synuclein and increased migratory capacity of microglia in PD, as well as the cooperation between microglia and exosomes in mediating α-synuclein release and propagation. In conclusion, this article help collate information on microglia in-relation to PD.
Collapse
Affiliation(s)
- Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China
| | - Zhengxiang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
23
|
Huynh B, Fu Y, Kirik D, Shine JM, Halliday GM. Comparison of Locus Coeruleus Pathology with Nigral and Forebrain Pathology in Parkinson's Disease. Mov Disord 2021; 36:2085-2093. [PMID: 33899954 DOI: 10.1002/mds.28615] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Pathology in the noradrenergic A6 locus coeruleus has not been compared with more rostral dopaminergic A9 substantia nigra and A10 ventral tegmental area, and cholinergic Ch4 basal nucleus and Ch1/2 septal regions in the same cases of Parkinson's disease (PD). OBJECTIVE To determine whether there is a gradient of caudal to rostral cell loss in PD. METHODS Postmortem brains were collected from longitudinally followed donors with PD (n = 14) and aged-matched healthy donors (n = 13), six with restricted brainstem Lewy pathology (RLP), fixed in formalin and serial tissue slabs processed for cell and pathological quantitation. Noradrenergic A6 neurons were assessed and compared with previously published midbrain and basal forebrain data. From these data, regression estimates of pathological onset and progression were determined. RESULTS Restricted Lewy pathology (RLP) cases had high pathological variability but no significant reduction in neurons. Pathology containing A6 neuron loss started at PD diagnosis and progressed faster (2.4% p.a) than the loss of dopaminergic A9 neurons (2% loss p.a.). Cases with dementia had significantly more pathology in noradrenergic and cholinergic neurons, had greater noradrenergic A6 neuron loss (29% more, progressing at 3.2% p.a.), and a selective loss of lateral A10 nonmelanized dopamine-producing neurons (starting a decade following diagnosis). CONCLUSIONS These findings show that in the same Parkinson's disease cases cell loss in these neurotransmitter systems does not follow a strict caudal to rostral trajectory and suggests symptom onset may relate to substantial pathology in the noradrenergic A6 locus coeruleus neurons in people with reduced dopamine-producing A9 substantia nigra neurons. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Benjamin Huynh
- ForeFront Research Team, Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuhong Fu
- ForeFront Research Team, Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Deniz Kirik
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - James M Shine
- ForeFront Research Team, Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | | |
Collapse
|
24
|
Brás IC, Outeiro TF. Alpha-Synuclein: Mechanisms of Release and Pathology Progression in Synucleinopathies. Cells 2021; 10:cells10020375. [PMID: 33673034 PMCID: PMC7917664 DOI: 10.3390/cells10020375] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The accumulation of misfolded alpha-synuclein (aSyn) throughout the brain, as Lewy pathology, is a phenomenon central to Parkinson’s disease (PD) pathogenesis. The stereotypical distribution and evolution of the pathology during disease is often attributed to the cell-to-cell transmission of aSyn between interconnected brain regions. The spreading of conformationally distinct aSyn protein assemblies, commonly referred as strains, is thought to result in a variety of clinically and pathologically heterogenous diseases known as synucleinopathies. Although tremendous progress has been made in the field, the mechanisms involved in the transfer of these assemblies between interconnected neural networks and their role in driving PD progression are still unclear. Here, we present an update of the relevant discoveries supporting or challenging the prion-like spreading hypothesis. We also discuss the importance of aSyn strains in pathology progression and the various putative molecular mechanisms involved in cell-to-cell protein release. Understanding the pathways underlying aSyn propagation will contribute to determining the etiology of PD and related synucleinopathies but also assist in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Inês C. Brás
- Center for Biostructural Imaging of Neurodegeneration, Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Tiago F. Outeiro
- Center for Biostructural Imaging of Neurodegeneration, Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with a Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
- Correspondence: ; Tel.: +49-(0)-551-391-3544; Fax: +49-(0)-551-392-2693
| |
Collapse
|
25
|
Levstek T, Redenšek S, Trošt M, Dolžan V, Podkrajšek KT. Assessment of the Telomere Length and Its Effect on the Symptomatology of Parkinson's Disease. Antioxidants (Basel) 2021; 10:antiox10010137. [PMID: 33478114 PMCID: PMC7835735 DOI: 10.3390/antiox10010137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 01/03/2023] Open
Abstract
Telomeres, which are repetitive sequences that cap the end of the chromosomes, shorten with each cell division. Besides cellular aging, there are several other factors that influence telomere length (TL), in particular, oxidative stress and inflammation, which play an important role in the pathogenesis of neurodegenerative brain diseases including Parkinson’s disease (PD). So far, the majority of studies have not demonstrated a significant difference in TL between PD patients and healthy individuals. However, studies investigating the effect of TL on the symptomatology and disease progression of PD are scarce, and thus, warranted. We analyzed TL of peripheral blood cells in a sample of 204 PD patients without concomitant autoimmune diseases and analyzed its association with several PD related phenotypes. Monochrome multiplex quantitative PCR (mmqPCR) was used to determine relative TL given as a ratio of the amount of DNA between the telomere and albumin as the housekeeping gene. We found a significant difference in the relative TL between PD patients with and without dementia, where shorter TL presented higher risk for dementia (p = 0.024). However, the correlation was not significant after adjustment for clinical factors (p = 0.509). We found no correlations between TLs and the dose of dopaminergic therapy when the analysis was adjusted for genetic variability in inflammatory or oxidative factors. In addition, TL influenced time to onset of motor complications after levodopa treatment initiation (p = 0.0134), but the association did not remain significant after adjustment for age at inclusion and disease duration (p = 0.0781). Based on the results of our study we conclude that TL contributes to certain PD-related phenotypes, although it may not have a major role in directing the course of the disease. Nevertheless, this expends currently limited knowledge regarding the association of the telomere attrition and the disease severity or motor complications in Parkinson’s disease.
Collapse
Affiliation(s)
- Tina Levstek
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (S.R.); (V.D.)
| | - Sara Redenšek
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (S.R.); (V.D.)
| | - Maja Trošt
- Department of Neurology, University Medical Centre Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia;
| | - Vita Dolžan
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (S.R.); (V.D.)
| | - Katarina Trebušak Podkrajšek
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (S.R.); (V.D.)
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Vrazov trg 1, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
26
|
Rastogi S, Sharma V, Bharti PS, Rani K, Modi GP, Nikolajeff F, Kumar S. The Evolving Landscape of Exosomes in Neurodegenerative Diseases: Exosomes Characteristics and a Promising Role in Early Diagnosis. Int J Mol Sci 2021; 22:E440. [PMID: 33406804 PMCID: PMC7795439 DOI: 10.3390/ijms22010440] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (ND) remains to be one of the biggest burdens on healthcare systems and serves as a leading cause of disability and death. Alzheimer's disease (AD) is among the most common of such disorders, followed by Parkinson's disease (PD). The basic molecular details of disease initiation and pathology are still under research. Only recently, the role of exosomes has been linked to the initiation and progression of these neurodegenerative diseases. Exosomes are small bilipid layer enclosed extracellular vesicles, which were once considered as a cellular waste and functionless. These nano-vesicles of 30-150 nm in diameter carry specific proteins, lipids, functional mRNAs, and high amounts of non-coding RNAs (miRNAs, lncRNAs, and circRNAs). As the exosomes content is known to vary as per their originating and recipient cells, these vesicles can be utilized as a diagnostic biomarker for early disease detection. Here we review exosomes, their biogenesis, composition, and role in neurodegenerative diseases. We have also provided details for their characterization through an array of available techniques. Their updated role in neurodegenerative disease pathology is also discussed. Finally, we have shed light on a novel field of salivary exosomes as a potential candidate for early diagnosis in neurodegenerative diseases and compared the biomarkers of salivary exosomes with other blood/cerebrospinal fluid (CSF) based exosomes within these neurological ailments.
Collapse
Affiliation(s)
- Simran Rastogi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Vaibhav Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Prahalad Singh Bharti
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Komal Rani
- Department of Biotechnology, Amity University, Mumbai 410206, India;
| | - Gyan P. Modi
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India;
| | - Fredrik Nikolajeff
- Department of Health Science, Lulea Technical University, 97187 Lulea, Sweden
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| |
Collapse
|
27
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
28
|
Otani K, Shichita T. Cerebral sterile inflammation in neurodegenerative diseases. Inflamm Regen 2020; 40:28. [PMID: 33292860 PMCID: PMC7722432 DOI: 10.1186/s41232-020-00137-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022] Open
Abstract
Therapeutic strategies for regulating neuroinflammation are expected in the development of novel therapeutic agents to prevent the progression of central nervous system (CNS) pathologies. An understanding of the detailed molecular and cellular mechanisms of neuroinflammation in each CNS disease is necessary for the development of therapeutics. Since the brain is a sterile organ, neuroinflammation in Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) is triggered by cerebral cellular damage or the abnormal accumulation of inflammatogenic molecules in CNS tissue through the activation of innate and acquired immunity. Inflammation and CNS pathologies worsen each other through various cellular and molecular mechanisms, such as oxidative stress or the accumulation of inflammatogenic molecules induced in the damaged CNS tissue. In this review, we summarize the recent evidence regarding sterile immune responses in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kento Otani
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-8512, Japan
| | - Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
- Precursory Research for Innovative Medical Care (PRIME), Japan Agency for Medical Research and Development (AMED), Tokyo, 100-0004, Japan.
| |
Collapse
|
29
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Patnaik R, Wiklund L, Sharma HS. Co-administration of TiO 2-nanowired dl-3-n-butylphthalide (dl-NBP) and mesenchymal stem cells enhanced neuroprotection in Parkinson's disease exacerbated by concussive head injury. PROGRESS IN BRAIN RESEARCH 2020; 258:101-155. [PMID: 33223034 DOI: 10.1016/bs.pbr.2020.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
dl-3-n-butylphthalide (dl-NBP) is a powerful antioxidant compound with profound neuroprotective effects in stroke and brain injury. However, its role in Parkinson's disease (PD) is not well known. Traumatic brain injury (TBI) is one of the key factors in precipitating PD like symptoms in civilians and particularly in military personnel. Thus, it would be interesting to explore the possible neuroprotective effects of NBP in PD following concussive head injury (CHI). In this chapter effect of nanowired delivery of NBP together with mesenchymal stem cells (MSCs) in PD with CHI is discussed based on our own investigations. It appears that CHI exacerbates PD pathophysiology in terms of p-tau, α-synuclein (ASNC) levels in the cerebrospinal fluid (CSF) and the loss of TH immunoreactivity in substantia niagra pars compacta (SNpc) and striatum (STr) along with dopamine (DA), dopamine decarboxylase (DOPAC). And homovanillic acid (HVA). Our observations are the first to show that a combination of NBP with MSCs when delivered using nanowired technology induces superior neuroprotective effects in PD brain pathology exacerbated by CHI, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, Hebei Province, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, Hebei Province, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
30
|
Heras-Garvin A, Stefanova N. From Synaptic Protein to Prion: The Long and Controversial Journey of α-Synuclein. Front Synaptic Neurosci 2020; 12:584536. [PMID: 33071772 PMCID: PMC7536368 DOI: 10.3389/fnsyn.2020.584536] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Since its discovery 30 years ago, α-synuclein (α-syn) has been one of the most studied proteins in the field of neuroscience. Dozens of groups worldwide have tried to reveal not only its role in the CNS but also in other organs. α-syn has been linked to several processes essential in brain homeostasis such as neurotransmitter release, synaptic function, and plasticity. However, despite the efforts made in this direction, the main function of α-syn is still unknown. Moreover, α-syn became a protein of interest for neurologists and neuroscientists when mutations in its gene were found associated with Parkinson's disease (PD) and even more when α-syn protein deposits were observed in the brain of PD, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) patients. At present, the abnormal accumulation of α-syn constitutes one of the pathological hallmarks of these disorders, also referred to as α-synucleinopathies, and it is used for post-mortem diagnostic criteria. Whether α-syn aggregation is cause or consequence of the pathogenic events underlying α-synucleinopathies remains unclear and under discussion. Recently, different in vitro and in vivo studies have shown the ability of pathogenic α-syn to spread between cells, not only within the CNS but also from peripheral locations such as the gut, salivary glands, and through the olfactory network into the CNS, inducing abnormal misfolding of endogenous α-syn and leading to neurodegeneration and motor and cognitive impairment in animal models. Thus, it has been suggested that α-syn should be considered a prion protein. Here we present an update of what we know about α-syn function, aggregation and spreading, and its role in neurodegeneration. We also discuss the rationale and findings supporting the hypothetical prion nature of α-syn, its weaknesses, and future perspectives for research and the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Antonio Heras-Garvin
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Schaser AJ, Stackhouse TL, Weston LJ, Kerstein PC, Osterberg VR, López CS, Dickson DW, Luk KC, Meshul CK, Woltjer RL, Unni VK. Trans-synaptic and retrograde axonal spread of Lewy pathology following pre-formed fibril injection in an in vivo A53T alpha-synuclein mouse model of synucleinopathy. Acta Neuropathol Commun 2020; 8:150. [PMID: 32859276 PMCID: PMC7456087 DOI: 10.1186/s40478-020-01026-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022] Open
Abstract
It is necessary to develop an understanding of the specific mechanisms involved in alpha-synuclein aggregation and propagation to develop disease modifying therapies for age-related synucleinopathies, including Parkinson's disease and Dementia with Lewy Bodies. To adequately address this question, we developed a new transgenic mouse model of synucleinopathy that expresses human A53T SynGFP under control of the mouse prion protein promoter. Our characterization of this mouse line demonstrates that it exhibits several distinct advantages over other, currently available, mouse models. This new model allows rigorous study of the initial location of Lewy pathology formation and propagation in the living brain, and strongly suggests that aggregation begins in axonal structures with retrograde propagation to the cell body. This model also shows expeditious development of alpha-synuclein pathology following induction with small, in vitro-generated alpha-synuclein pre-formed fibrils (PFFs), as well as accelerated cell death of inclusion-bearing cells. Using this model, we found that aggregated alpha-synuclein somatic inclusions developed first in neurons, but later showed a second wave of inclusion formation in astrocytes. Interestingly, astrocytes appear to survive much longer after inclusion formation than their neuronal counterparts. This model also allowed careful study of peripheral-to-central spread of Lewy pathology after PFF injection into the hind limb musculature. Our results clearly show evidence of progressive, retrograde trans-synaptic spread of Lewy pathology through known neuroanatomically connected pathways in the motor system. As such, we have developed a promising tool to understand the biology of neurodegeneration associated with alpha-synuclein aggregation and to discover new treatments capable of altering the neurodegenerative disease course of synucleinopathies.
Collapse
Affiliation(s)
- Allison J Schaser
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Teresa L Stackhouse
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Leah J Weston
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Patrick C Kerstein
- Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Valerie R Osterberg
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Claudia S López
- Multiscale Microscopy Core, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Charles K Meshul
- Research Services, Veterans Affairs Medical Center, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Randall L Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Vivek K Unni
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, 97239, USA.
- Parkinson Center, Department of Neurology, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
32
|
Kaniyappan S, Tepper K, Biernat J, Chandupatla RR, Hübschmann S, Irsen S, Bicher S, Klatt C, Mandelkow EM, Mandelkow E. FRET-based Tau seeding assay does not represent prion-like templated assembly of Tau filaments. Mol Neurodegener 2020; 15:39. [PMID: 32677995 PMCID: PMC7364478 DOI: 10.1186/s13024-020-00389-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/08/2020] [Indexed: 01/15/2023] Open
Abstract
Tau aggregation into amyloid fibers based on the cross-beta structure is a hallmark of several Tauopathies, including Alzheimer Disease (AD). Trans-cellular propagation of Tau with pathological conformation has been suggested as a key disease mechanism. This is thought to cause the spreading of Tau pathology in AD by templated conversion of naive Tau in recipient cells into a pathological state, followed by assembly of pathological Tau fibers, similar to the mechanism of nucleated polymerization proposed for prion pathogenesis. In cell cultures, the process is often monitored by a FRET assay where the recipient cell expresses the Tau repeat domain (TauRD) with a pro-aggregant mutation, fused to GFP-based FRET pairs. Since the size of the reporter GFP (barrel of ~ 3 nm × 4 nm) is ~ 7 times larger than the β-strand distance (0.47 nm), this points to a potential steric clash. Hence, we investigated the influence of the GFP tag on TauFL or TauRD aggregation. Using biophysical methods (light scattering, atomic force microscopy (AFM), and scanning-transmission electron microscopy (STEM)), we found that the assembly of TauRD-GFP was severely inhibited and incompatible with that of Alzheimer filaments. These observations argue against the hypothesis that the propagation of Tau pathology in AD is caused by the prion-like templated aggregation of Tau protein, transmitted via cell-to-cell spreading of Tau. Thus, even though the observed local increase of FRET in recipient cells may be a valid hallmark of a pathological reaction, our data argue that it is caused by a process distinct from assembly of TauRD filaments.
Collapse
Affiliation(s)
- Senthilvelrajan Kaniyappan
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany. .,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.
| | - Katharina Tepper
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Jacek Biernat
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | | | | | | | | | - Eva-Maria Mandelkow
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany.,CAESAR Research Center, Bonn, Germany
| | - Eckhard Mandelkow
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany. .,CAESAR Research Center, Bonn, Germany.
| |
Collapse
|
33
|
Ásgrímsdóttir ES, Arenas E. Midbrain Dopaminergic Neuron Development at the Single Cell Level: In vivo and in Stem Cells. Front Cell Dev Biol 2020; 8:463. [PMID: 32733875 PMCID: PMC7357704 DOI: 10.3389/fcell.2020.00463] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that predominantly affects dopaminergic (DA) neurons of the substantia nigra. Current treatment options for PD are symptomatic and typically involve the replacement of DA neurotransmission by DA drugs, which relieve the patients of some of their motor symptoms. However, by the time of diagnosis, patients have already lost about 70% of their substantia nigra DA neurons and these drugs offer only temporary relief. Therefore, cell replacement therapy has garnered much interest as a potential treatment option for PD. Early studies using human fetal tissue for transplantation in PD patients provided proof of principle for cell replacement therapy, but they also highlighted the ethical and practical difficulties associated with using human fetal tissue as a cell source. In recent years, advancements in stem cell research have made human pluripotent stem cells (hPSCs) an attractive source of material for cell replacement therapy. Studies on how DA neurons are specified and differentiated in the developing mouse midbrain have allowed us to recapitulate many of the positional and temporal cues needed to generate DA neurons in vitro. However, little is known about the developmental programs that govern human DA neuron development. With the advent of single-cell RNA sequencing (scRNA-seq) and bioinformatics, it has become possible to analyze precious human samples with unprecedented detail and extract valuable high-quality information from large data sets. This technology has allowed the systematic classification of cell types present in the human developing midbrain along with their gene expression patterns. By studying human development in such an unbiased manner, we can begin to elucidate human DA neuron development and determine how much it differs from our knowledge of the rodent brain. Importantly, this molecular description of the function of human cells has become and will increasingly be a reference to define, evaluate, and engineer cell types for PD cell replacement therapy and disease modeling.
Collapse
Affiliation(s)
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Role of Long Noncoding RNAs in Parkinson's Disease: Putative Biomarkers and Therapeutic Targets. PARKINSONS DISEASE 2020; 2020:5374307. [PMID: 32617144 PMCID: PMC7306067 DOI: 10.1155/2020/5374307] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/07/2020] [Accepted: 05/21/2020] [Indexed: 01/12/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by bradykinesia, rigidity, and tremor. Age is the main risk factor. Long noncoding RNAs (lncRNAs) are novel RNA molecules of more than 200 nucleotides in length. They may be involved in the regulation of many pathological processes of PD. PD has a variety of pathophysiological mechanisms, including alpha-synuclein aggregate, mitochondrial dysfunction, oxidative stress, calcium homeostasis, axonal transport, and neuroinflammation. Among these, the impacts of lncRNAs on the pathogenesis and progression of PD need to be highlighted. lncRNAs may serve as putative biomarkers and therapeutic targets for the early diagnosis of PD. This study aimed to investigate the role of lncRNAs in various pathological processes of PD and the specific lncRNAs that might be used as putative diagnostic biomarkers and therapeutic targets of PD.
Collapse
|
35
|
Janitzky K. Impaired Phasic Discharge of Locus Coeruleus Neurons Based on Persistent High Tonic Discharge-A New Hypothesis With Potential Implications for Neurodegenerative Diseases. Front Neurol 2020; 11:371. [PMID: 32477246 PMCID: PMC7235306 DOI: 10.3389/fneur.2020.00371] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/14/2020] [Indexed: 12/21/2022] Open
Abstract
The locus coeruleus (LC) is a small brainstem nucleus with widely distributed noradrenergic projections to the whole brain, and loss of LC neurons is a prominent feature of age-related neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). This article discusses the hypothesis that in early stages of neurodegenerative diseases, the discharge mode of LC neurons could be changed to a persistent high tonic discharge, which in turn might impair phasic discharge. Since phasic discharge of LC neurons is required for the release of high amounts of norepinephrine (NE) in the brain to promote anti-inflammatory and neuroprotective effects, persistent high tonic discharge of LC neurons could be a key factor in the progression of neurodegenerative diseases. Transcutaneous vagal stimulation (t-VNS), a non-invasive technique that potentially increases phasic discharge of LC neurons, could therefore provide a non-pharmacological treatment approach in specific disease stages. This article focuses on LC vulnerability in neurodegenerative diseases, discusses the hypothesis that a persistent high tonic discharge of LC neurons might affect neurodegenerative processes, and finally reflects on t-VNS as a potentially useful clinical tool in specific stages of AD and PD.
Collapse
Affiliation(s)
- Kathrin Janitzky
- Department of Neurology, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
36
|
Meng L, Yuan X, Cao X, Zhang Z. The gut-brain axis in the pathogenesis of Parkinson’s disease. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.1177/2096595820902566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. Its pathological markers include Lewy bodies and Lewy neuritis, which primarily affect the substantia nigra. However, in recent years, mounting evidence suggests that PD is a multifocal neurodegenerative process that influences several neuronal structures aside from the substantia nigra, one of which is the enteric nervous system. Many clinical studies have reported that patients with PD experience gastrointestinal dysfunction for many years before the onset of motor symptoms. Emerging evidence indicates that α-synuclein deposition may start in the enteric nervous system and then propagate to the central nervous system. The gut-brain axis plays an important role in PD pathogenesis. Recent evidence suggests that these interactions may be primarily affected by the intestinal microbiota. In this review, the authors discuss recent research, and illustrate how changes in the composition of the gut microbiota may trigger inflammation, thus contributing to neurodegeneration in PD.
Collapse
Affiliation(s)
- Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- These authors contributed equally to this work
| | - Xin Yuan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- These authors contributed equally to this work
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
37
|
Jung YJ, Tweedie D, Scerba MT, Greig NH. Neuroinflammation as a Factor of Neurodegenerative Disease: Thalidomide Analogs as Treatments. Front Cell Dev Biol 2019; 7:313. [PMID: 31867326 PMCID: PMC6904283 DOI: 10.3389/fcell.2019.00313] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation is initiated when glial cells, mainly microglia, are activated by threats to the neural environment, such as pathogen infiltration or neuronal injury. Although neuroinflammation serves to combat these threats and reinstate brain homeostasis, chronic inflammation can result in excessive cytokine production and cell death if the cause of inflammation remains. Overexpression of tumor necrosis factor-α (TNF-α), a proinflammatory cytokine with a central role in microglial activation, has been associated with neuronal excitotoxicity, synapse loss, and propagation of the inflammatory state. Thalidomide and its derivatives, termed immunomodulatory imide drugs (IMiDs), are a class of drugs that target the 3'-untranslated region (3'-UTR) of TNF-α mRNA, inhibiting TNF-α production. Due to their multi-potent effects, several IMiDs, including thalidomide, lenalidomide, and pomalidomide, have been repurposed as drug treatments for diseases such as multiple myeloma and psoriatic arthritis. Preclinical studies of currently marketed IMiDs, as well as novel IMiDs such as 3,6'-dithiothalidomide and adamantyl thalidomide derivatives, support the development of IMiDs as therapeutics for neurological disease. IMiDs have a competitive edge compared to similar anti-inflammatory drugs due to their blood-brain barrier permeability and high bioavailability, with the potential to alleviate symptoms of neurodegenerative disease and slow disease progression. In this review, we evaluate the role of neuroinflammation in neurodegenerative diseases, focusing specifically on the role of TNF-α in neuroinflammation, as well as appraise current research on the potential of IMiDs as treatments for neurological disorders.
Collapse
Affiliation(s)
- Yoo Jin Jung
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | | | | | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
38
|
Mezias C, Rey N, Brundin P, Raj A. Neural connectivity predicts spreading of alpha-synuclein pathology in fibril-injected mouse models: Involvement of retrograde and anterograde axonal propagation. Neurobiol Dis 2019; 134:104623. [PMID: 31628991 PMCID: PMC7138530 DOI: 10.1016/j.nbd.2019.104623] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/05/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
In Parkinson’s disease, some of the first alpha-synuclein aggregates appear in the olfactory system and the dorsal motor nucleus of the vagus nerve before spreading to connected brain regions. We previously demonstrated that injection of alpha-synuclein fibrils unilaterally into the olfactory bulb of wild type mice leads to widespread synucleinopathy in brain regions directly and indirectly connected to the injection site, consistently, over the course of periods longer than 6 months. Our previously reported observations support the idea that alpha-synuclein inclusions propagates between brain region through neuronal networks. In the present study, we further defined the pattern of propagation of alpha-synuclein inclusions and developed a mathematical model based on known mouse brain connectivity. Using this model, we first predicted the pattern of alpha-synuclein inclusions propagation following an injection of fibrils into the olfactory bulb. We then analyzed the fitting of these predictions to our published histological data. Our results demonstrate that the pattern of propagation we observed in vivo is consistent with axonal transport of alpha-synuclein aggregate seeds, followed by transsynaptic transmission. By contrast, simple diffusion of alpha-synuclein fits very poorly our in vivo data. We also found that the spread of alpha-synuclein inclusions appeared to primarily follow neural connections retrogradely until 9 months after injection into the olfactory bulb. Thereafter, the pattern of spreading was consistent with anterograde propagation mathematical models. Finally, we applied our mathematical model to a different, previously published, dataset involving alpha-synuclein fibril injections into the striatum, instead of the olfactory bulb. We found that the mathematical model accurately predicts the reported progressive increase in alpha-synuclein neuropathology also in that paradigm. In conclusion, our findings support that the progressive spread of alpha-synuclein inclusions after injection of protein fibrils follows neural networks in the mouse connectome.
Collapse
Affiliation(s)
- Christopher Mezias
- Weill Cornell Medicine of Cornell University, Department of Neuroscience, New York, NY 10065, USA; Weill Cornell Medicine of Cornell University, Department of Radiology, New York, NY 10065, USA.
| | - Nolwen Rey
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ashish Raj
- Weill Cornell Medicine of Cornell University, Department of Radiology, New York, NY 10065, USA; University of California-San Francisco, Department of Biomedical Imaging, USA
| |
Collapse
|
39
|
Affiliation(s)
- Heiko Braak
- Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany.
| | - Kelly Del Tredici-Braak
- Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076, Tübingen, Germany
| |
Collapse
|
40
|
Porro C, Panaro MA, Lofrumento DD, Hasalla E, Trotta T. The multiple roles of exosomes in Parkinson's disease: an overview. Immunopharmacol Immunotoxicol 2019; 41:469-476. [PMID: 31405314 DOI: 10.1080/08923973.2019.1650371] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extracellular vesicles (EVs) represent a relatively new field of research in neurodegenerative disease and they are thought to be one of the ways that neurodegenerative pathologies, such as Parkinson's Disease (PD), spread in the brain. EVs are membrane vesicles released from cells into the extracellular space and they are produced by all cells of the nervous tissue. The classification of the vesicle subtypes comprises exosomes, microvesicles/microparticles, apoptotic bodies. EVs change in number and content in response to environmental conditions and may function as shuttles for the delivery of cargo between cells. Recent data suggest that exosomes secreted by both activated microglia and neurons play an important role in α-synuclein (α-syn) spreading and increase of neuroinflammation, thus exacerbating neuronal dysfunction and disease progression. α-syn is a presynaptic protein secreted by neurons in small amounts, and it is the main component of Lewy bodies, one of the histopathological features of PD. Several factors have shown to induce and/or modulate α-syn structure and oligomerization in vitro. Under pathological conditions, progressive accumulation of α-syn and the formation of oligomers have been proposed to play a critical role in the pathogenesis of PD. This review gives an overview about the multiple roles of exosomes in PD, despite their role in the progression of neurodegeneration, exosomes could represent a specific drug delivery tool for a difficult target such as the brain, which poses an obstacle to most drugs and they could also represent new biomarkers to track the progression of PD.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari , Italy
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento , Lecce , Italy
| | - Elona Hasalla
- Department of Pre-Clinic Subjects, Faculty of Medical Sciences, University of Elbasan "Aleksander Xhuvani" , Elbasan , Albania
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| |
Collapse
|
41
|
Meng L, Yuan X, Cao X, Zhang Z. The gut-brain axis in the pathogenesis of Parkinson’s disease. BRAIN SCIENCE ADVANCES 2019. [DOI: 10.26599/bsa.2019.9050009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
42
|
Flores-Cuadrado A, Saiz-Sanchez D, Mohedano-Moriano A, Martinez-Marcos A, Ubeda-Bañon I. Neurodegeneration and contralateral α-synuclein induction after intracerebral α-synuclein injections in the anterior olfactory nucleus of a Parkinson's disease A53T mouse model. Acta Neuropathol Commun 2019; 7:56. [PMID: 30987677 PMCID: PMC6463651 DOI: 10.1186/s40478-019-0713-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease is characterized by a proteinopathy that includes aggregates of α-synuclein. A recent hypothesis proposes a prion-like spreading mechanism for this α-synucleinopathy. Early neuropathological deposits occur, among others, in the anterior olfactory nucleus (AON). This study investigates the anterograde and/or retrograde transmissibility of exogenous α-synuclein inoculated in the right AON of the A53T model of Parkinson’s disease and wild-type mice as well as neuronal and glial involvement. Seven experimental groups were established: wild-type injected with tracers; A53T mice injected with either α-synuclein or saline 2 months beforehand; wild-type injected with either α-synuclein or saline 2 months beforehand; and wild-type injected with either α-synuclein or saline 4 months beforehand. Weight and behavioral changes were analyzed. Immunohistochemistry against α-synuclein, NeuN, Iba-1 and GFAP was performed. Volume and marker distributions in the olfactory bulb (OB), AON and piriform cortex were analyzed using unbiased stereology. The behavioral analyses reveal higher levels of hyperactivity in transgenic as compared to wild-type mice. Tract-tracing experiments show that the main contralateral afferent projections to the dorsal AON come from the AON and secondarily from the OB. In saline-injected transgenic animals, α-synuclein expression in the OB and the AON is higher in the left hemisphere than in the right hemisphere, which could be due to basal interhemispheric differences. α-synuclein injection could provoke a significant increase in the left hemisphere of the transgenic mice’s OB, compared to saline-injected animals. Neuronal loss was observed in saline-injected transgenic mice relative to the saline-injected wild-type group. There were no overall differences in neuron number following injection of α-synuclein into either wild-type or transgenic mice, however some neuron loss was apparent in specific regions of α-synuclein injected wild-types. Microglia labeling appeared to be correlated with surgery-induced inflammation. Astroglial labeling was higher in transgenic animals, which could be due to endogenous α-synucleinopathy. This study suggests α-synucleinopathy induction, via retrograde and contralateral projections, within the olfactory system of transgenic animals.
Collapse
|
43
|
Outeiro TF, Koss DJ, Erskine D, Walker L, Kurzawa-Akanbi M, Burn D, Donaghy P, Morris C, Taylor JP, Thomas A, Attems J, McKeith I. Dementia with Lewy bodies: an update and outlook. Mol Neurodegener 2019; 14:5. [PMID: 30665447 PMCID: PMC6341685 DOI: 10.1186/s13024-019-0306-8] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is an age-associated neurodegenerative disorder producing progressive cognitive decline that interferes with normal life and daily activities. Neuropathologically, DLB is characterised by the accumulation of aggregated α-synuclein protein in Lewy bodies and Lewy neurites, similar to Parkinson’s disease (PD). Extrapyramidal motor features characteristic of PD, are common in DLB patients, but are not essential for the clinical diagnosis of DLB. Since many PD patients develop dementia as disease progresses, there has been controversy about the separation of DLB from PD dementia (PDD) and consensus reports have put forward guidelines to assist clinicians in the identification and management of both syndromes. Here, we present basic concepts and definitions, based on our current understanding, that should guide the community to address open questions that will, hopefully, lead us towards improved diagnosis and novel therapeutic strategies for DLB and other synucleinopathies.
Collapse
Affiliation(s)
- Tiago Fleming Outeiro
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK. .,Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany. .,Max Planck Institute for Experimental Medicine, Göttingen, Germany.
| | - David J Koss
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Daniel Erskine
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Lauren Walker
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Marzena Kurzawa-Akanbi
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - David Burn
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Paul Donaghy
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Christopher Morris
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - John-Paul Taylor
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Alan Thomas
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Johannes Attems
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Ian McKeith
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
44
|
Brundin P, Coetzee GA. Genetically engineered stem cell-derived neurons can be rendered resistant to alpha-synuclein aggregate pathology. Eur J Neurosci 2019; 49:316-319. [PMID: 30614081 DOI: 10.1111/ejn.14333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| | - Gerhard A Coetzee
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|
45
|
Brás IC, Lopes LV, Outeiro TF. Sensing α-Synuclein From the Outside via the Prion Protein: Implications for Neurodegeneration. Mov Disord 2018; 33:1675-1684. [DOI: 10.1002/mds.27478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; Lisboa Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciências Médicas; Universidade Nova de Lisboa, Campo dos Mártires da Pátria; Lisboa Portugal
- Max Planck Institute for Experimental Medicine; Göttingen Germany
- Institute of Neuroscience, The Medical School; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
46
|
Sang J, Meisl G, Thackray AM, Hong L, Ponjavic A, Knowles TPJ, Bujdoso R, Klenerman D. Direct Observation of Murine Prion Protein Replication in Vitro. J Am Chem Soc 2018; 140:14789-14798. [PMID: 30351023 PMCID: PMC6225343 DOI: 10.1021/jacs.8b08311] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Prions are believed to propagate when an assembly of prion protein (PrP) enters a cell and replicates to produce two or more fibrils, leading to an exponential increase in PrP aggregate number with time. However, the molecular basis of this process has not yet been established in detail. Here, we use single-aggregate imaging to study fibril fragmentation and elongation of individual murine PrP aggregates from seeded aggregation in vitro. We found that PrP elongation occurs via a structural conversion from a PK-sensitive to PK-resistant conformer. Fibril fragmentation was found to be length-dependent and resulted in the formation of PK-sensitive fragments. Measurement of the rate constants for these processes also allowed us to predict a simple spreading model for aggregate propagation through the brain, assuming that doubling of the aggregate number is rate-limiting. In contrast, while α-synuclein aggregated by the same mechanism, it showed significantly slower elongation and fragmentation rate constants than PrP, leading to much slower replication rate. Overall, our study shows that fibril elongation with fragmentation are key molecular processes in PrP and α-synuclein aggregate replication, an important concept in prion biology, and also establishes a simple framework to start to determine the main factors that control the rate of prion and prion-like spreading in animals.
Collapse
Affiliation(s)
- Jason
C. Sang
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.
| | - Georg Meisl
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.
| | - Alana M. Thackray
- Department
of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES, U.K.
| | - Liu Hong
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.,Zhou
Pei-Yuan Center for Applied Mathematics, Tsinghua University, Beijing 100084, PR China
| | - Aleks Ponjavic
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.
| | - Tuomas P. J. Knowles
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.,Cavendish
Laboratory, University of Cambridge, Cambridge, CB3 0HE, U.K.
| | - Raymond Bujdoso
- Department
of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES, U.K.
| | - David Klenerman
- Department
of Chemistry, University of Cambridge, Cambridge, CB2 1EW, U.K.,
| |
Collapse
|
47
|
Non-cell-autonomous actions of α-synuclein: Implications in glial synucleinopathies. Prog Neurobiol 2018; 169:158-171. [DOI: 10.1016/j.pneurobio.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/24/2017] [Accepted: 06/30/2018] [Indexed: 01/11/2023]
|
48
|
Uemura N, Yagi H, Uemura MT, Hatanaka Y, Yamakado H, Takahashi R. Inoculation of α-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol Neurodegener 2018; 13:21. [PMID: 29751824 PMCID: PMC5948849 DOI: 10.1186/s13024-018-0257-5] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Intraneuronal α-synuclein (α-Syn) aggregates known as Lewy bodies (LBs) and the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) are the pathological hallmarks of Parkinson's disease (PD). Braak's hypothesis based on autopsy studies suggests that Lewy pathology initially occurs in the enteric nervous system (ENS) and then travels retrogradely to the dorsal motor nucleus of the vagus nerve (dmX), proceeding from there in a caudo-rostral direction. Recent evidence that α-Syn aggregates propagate between interconnected neurons supports this hypothesis. However, there is no direct evidence demonstrating this transmission from the ENS to the dmX and then to the SNpc. METHODS We inoculated α-Syn preformed fibrils (PFFs) or phosphate-buffered saline (PBS) into the mouse gastric wall and analyzed the progression of the pathology. RESULTS The mice inoculated with α-Syn PFFs, but not with PBS, developed phosphorylated α-Syn (p-α-Syn)-positive LB-like aggregates in the dmX at 45 days postinoculation. This aggregate formation was completely abolished when vagotomy was performed prior to inoculation of α-Syn PFFs, suggesting that the aggregates in the dmX were retrogradely induced via the vagus nerve. Unexpectedly, the number of neurons containing p-α-Syn-positive aggregates in the dmX decreased over time, and no further caudo-rostral propagation beyond the dmX was observed up to 12 months postinoculation. P-α-Syn-positive aggregates were also present in the myenteric plexus at 12 months postinoculation. However, unlike in patients with PD, there was no cell-type specificity in neurons containing those aggregates in this model. CONCLUSIONS These results indicate that α-Syn PFF inoculation into the mouse gastrointestinal tract can induce α-Syn pathology resembling that of very early PD, but other factors are apparently required if further progression of PD pathology is to be replicated in this animal model.
Collapse
Affiliation(s)
- Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Kyoto, Sakyoku, 606-8507, Japan.
| | - Hisashi Yagi
- Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyamacho-minami, Tottori, Tottori, 680-8550, Japan
| | - Maiko T Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Kyoto, Sakyoku, 606-8507, Japan
| | - Yusuke Hatanaka
- Department of Neurology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Kyoto, Sakyoku, 606-8507, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Kyoto, Sakyoku, 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Kyoto, Sakyoku, 606-8507, Japan.
| |
Collapse
|
49
|
Yan Z, Gibson SA, Buckley JA, Qin H, Benveniste EN. Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin Immunol 2018; 189:4-13. [PMID: 27713030 PMCID: PMC5573639 DOI: 10.1016/j.clim.2016.09.014] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023]
Abstract
The Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) signaling pathway is utilized by numerous cytokines and interferons, and is essential for the development and function of both innate and adaptive immunity. Aberrant activation of the JAK/STAT pathway is evident in neuroinflammatory diseases such as Multiple Sclerosis and Parkinson's Disease. Innate immunity is the front line defender of the immune system and is composed of various cell types, including microglia, macrophages and neutrophils. Innate immune responses have both pathogenic and protective roles in neuroinflammation, depending on disease context and the microenvironment in the central nervous system. In this review, we discuss the role of innate immunity in the pathogenesis of neuroinflammatory diseases, how the JAK/STAT signaling pathway regulates the innate immune response, and finally, the potential for ameliorating neuroinflammation by utilization of JAK/STAT inhibitors.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jessica A Buckley
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
50
|
Steiner JA, Quansah E, Brundin P. The concept of alpha-synuclein as a prion-like protein: ten years after. Cell Tissue Res 2018; 373:161-173. [PMID: 29480459 DOI: 10.1007/s00441-018-2814-1] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/29/2018] [Indexed: 12/18/2022]
Abstract
Parkinson's disease is characterized by the loss of nigrostriatal dopaminergic signaling and the presence of alpha-synuclein aggregates (also called Lewy bodies and neurites) throughout the brain. In 2003, Braak and colleagues created a staging system for Parkinson's disease describing the connection between the alpha-synuclein pathology and disease severity. Later, they suggested that the pathology might initially be triggered by exogenous insults targeting the gut and olfactory system. In 2008, we and other groups documented Lewy pathology in grafted neurons in people with Parkinson's disease who had been transplanted over a decade prior to autopsy. We proposed that the Lewy pathology in the grafted neurons was the result of permissive templating or prion-like spread of alpha-synuclein pathology from neurons in the host to those in the grafts. During the following ten years, several studies described the transmission of alpha-synuclein pathology between neurons, both in cell culture and in experimental animals. Recent research has also begun to identify underlying molecular mechanisms. Collectively, these experimental studies tentatively support the idea that the progression from one Braak stage to the next is the consequence of prion-like propagation of Lewy pathology. However, definitive proof that intercellular propagation of alpha-synuclein pathology occurs in Parkinson's disease cases has proven difficult to secure. In this review, we highlight several open questions that currently prevent us from concluding with certainty that prion-like transfer of alpha-synuclein contributes to the progression of Parkinson's disease.
Collapse
Affiliation(s)
- Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave. NE, Grand Rapids, MI, 49503, USA.
| | - Emmanuel Quansah
- Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave. NE, Grand Rapids, MI, 49503, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave. NE, Grand Rapids, MI, 49503, USA
| |
Collapse
|