1
|
Wang Y, Chen R, Shi G, Huang X, Li K, Wang R, Cao X, Yang Z, Zhao N, Yan J. Chitosan alleviates symptoms of Parkinson's disease by reducing acetate levels, which decreases inflammation and promotes repair of the intestinal barrier and blood-brain barrier. Neural Regen Res 2024; 21:01300535-990000000-00398. [PMID: 38934394 PMCID: PMC12094542 DOI: 10.4103/nrr.nrr-d-23-01511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/02/2023] [Accepted: 01/17/2024] [Indexed: 06/28/2024] Open
Abstract
ABSTRACT Studies have shown that chitosan protects against neurodegenerative diseases. However, the precise mechanism remains poorly understood. In this study, we administered chitosan intragastrically to an MPTP-induced mouse model of Parkinson's disease and found that it effectively reduced dopamine neuron injury, neurotransmitter dopamine release, and motor symptoms. These neuroprotective effects of chitosan were related to bacterial metabolites, specifically short-chain fatty acids, and chitosan administration altered intestinal microbial diversity and decreased short-chain fatty acid production in the gut. Furthermore, chitosan effectively reduced damage to the intestinal barrier and the blood-brain barrier. Finally, we demonstrated that chitosan improved intestinal barrier function and alleviated inflammation in both the peripheral nervous system and the central nervous system by reducing acetate levels. Based on these findings, we suggest a molecular mechanism by which chitosan decreases inflammation through reducing acetate levels and repairing the intestinal and blood-brain barriers, thereby alleviating symptoms of Parkinson's disease.
Collapse
Affiliation(s)
- Yinying Wang
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
- Yunnan Provincial Key Laboratory of Molecular Biology for Sino Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan Province, China
| | - Rongsha Chen
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Guolin Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xinwei Huang
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Ke Li
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Ruohua Wang
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xia Cao
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sino Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan Province, China
| | - Ninghui Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jinyuan Yan
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
2
|
Huang J, Li W. Molecular crosstalk between circadian clock and NLRP3 inflammasome signaling in Parkinson's disease. Heliyon 2024; 10:e24752. [PMID: 38268831 PMCID: PMC10803942 DOI: 10.1016/j.heliyon.2024.e24752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Research has recently found that both animal models and patients with PD have circadian dysfunction, accompanied by abnormal expression of circadian genes and proteins, which implies that the circadian clock plays a crucial role in PD etiopathogenesis. In addition, a strong relationship between NLRP3 inflammasome signaling and PD has been observed. Meanwhile, the activation of the NLRP3 inflammasome is highly relevant to dysfunctions of the molecular clock. Therefore, alleviating the neuroinflammation caused by NLRP3 inflammasome signaling by adjusting the abnormal molecular clock may be a potential strategy for preventing and treating PD. In this article, we have reviewed the potential or direct relationship between abnormalities of the circadian clock and NLRP3 inflammasome signaling in PD.
Collapse
Affiliation(s)
- Jiahua Huang
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| | - Wenwei Li
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| |
Collapse
|
3
|
Yu X, Jia Y, Dong Y. Research progress on the cannabinoid type-2 receptor and Parkinson's disease. Front Aging Neurosci 2024; 15:1298166. [PMID: 38264546 PMCID: PMC10804458 DOI: 10.3389/fnagi.2023.1298166] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
Parkinson's disease (PD) is featured by movement impairments, including tremors, bradykinesia, muscle stiffness, and imbalance. PD is also associated with many non-motor symptoms, such as cognitive impairments, dementia, and mental disorders. Previous studies identify the associations between PD progression and factors such as α-synuclein aggregation, mitochondrial dysfunction, inflammation, and cell death. The cannabinoid type-2 receptor (CB2 receptor) is a transmembrane G-protein-coupled receptor and has been extensively studied as part of the endocannabinoid system. CB2 receptor is recently emerged as a promising target for anti-inflammatory treatment for neurodegenerative diseases. It is reported to modulate mitochondrial function, oxidative stress, iron transport, and neuroinflammation that contribute to neuronal cell death. Additionally, CB2 receptor possesses the potential to provide feedback on electrophysiological processes, offering new possibilities for PD treatment. This review summarized the mechanisms underlying PD pathogenesis. We also discussed the potential regulatory role played by CB2 receptor in PD.
Collapse
Affiliation(s)
- Xiaoqi Yu
- Neuropsychiatry Research Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Yi Jia
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Yuan Dong
- Neuropsychiatry Research Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Huang J, Zhang X, Yang X, Yv Q, Ye F, Chen S, Cui Y, Gu L, Zhu M, Li W. Baicalin exerts neuroprotective actions by regulating the Nrf2-NLRP3 axis in toxin-induced models of Parkinson's disease. Chem Biol Interact 2024; 387:110820. [PMID: 38016618 DOI: 10.1016/j.cbi.2023.110820] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Baicalin, a potent anti-oxidative and anti-inflammatory flavonoid compound derived from Scutellaria baicalensis, has emerged as a neuroprotective agent. However, the mechanisms by which baicalin is neuroprotective in Parkinson's disease (PD) remain unclear. In this research, α-syn/MPP+ and MPTP were used to establish PD models in BV2 cells and C57BL/6 mice, respectively. The effect and mechanism of action of baicalin in PD were investigated by Western blotting, RT-qPCR, ELISA, Immunohistochemistry (IHC) staining, Immunofluorescence (IF) staining, HPLC and methods. Results demonstrate that baicalin mitigates oxidative stress, microglia activation and inflammatory response caused by α-syn/MPP+ and MPTP. It protects against dopaminergic neuron loss and relieves motor deficits. Meanwhile, baicalin not only significantly up-regulates the expression of Nrf2 and its downstream antioxidant enzyme, but also suppresses the activation of NLRP3 inflammasome simultaneously. Notably, the beneficial effects of baicalin in PD treatment are blocked by Nrf2 knockdown. This research reveals that baicalin may exert neuroprotective effects in PD treatment by suppressing the activation of NLRP3 inflammasome and it is dependent on the Nrf2-mediated antioxidative response.
Collapse
Affiliation(s)
- Jiahua Huang
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xueping Yang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qingyun Yv
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Fanlong Ye
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Sheng Chen
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Ying Cui
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Linting Gu
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China
| | - Min Zhu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
| | - Wenwei Li
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201500, China; Institute of Neurology, Institutes of Integrative Medicine, Fudan University, Shanghai 201500, China.
| |
Collapse
|
5
|
Boyd SL, Kuhn NC, Patterson JR, Stoll AC, Zimmerman SA, Kolanowski MR, Neubecker JJ, Luk KC, Ramsson ES, Sortwell CE, Bernstein AI. Developmental exposure to the Parkinson's disease-associated organochlorine pesticide dieldrin alters dopamine neurotransmission in α-synuclein pre-formed fibril (PFF)-injected mice. Toxicol Sci 2023; 196:99-111. [PMID: 37607008 PMCID: PMC10613968 DOI: 10.1093/toxsci/kfad086] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023] Open
Abstract
Parkinson's disease (PD) is the fastest-growing neurological disease worldwide, with increases outpacing aging and occurring most rapidly in recently industrialized areas, suggesting a role of environmental factors. Epidemiological, post-mortem, and mechanistic studies suggest that persistent organic pollutants, including the organochlorine pesticide dieldrin, increase PD risk. In mice, developmental dieldrin exposure causes male-specific exacerbation of neuronal susceptibility to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and synucleinopathy. Specifically, in the α-synuclein (α-syn) pre-formed fibril (PFF) model, exposure leads to increased deficits in striatal dopamine (DA) turnover and motor deficits on the challenging beam. Here, we hypothesized that alterations in DA handling contribute to the observed changes and assessed vesicular monoamine transporter 2 (VMAT2) function and DA release in this dieldrin/PFF 2-hit model. Female C57BL/6 mice were exposed to 0.3 mg/kg dieldrin or vehicle every 3 days by feeding, starting at 8 weeks of age and continuing throughout breeding, gestation, and lactation. Male offspring from independent litters underwent unilateral, intrastriatal injections of α-syn PFFs at 12 weeks of age, and vesicular 3H-DA uptake assays and fast-scan cyclic voltammetry were performed 4 months post-PFF injection. Dieldrin-induced an increase in DA release in striatal slices in PFF-injected animals, but no change in VMAT2 activity. These results suggest that developmental dieldrin exposure increases a compensatory response to synucleinopathy-triggered striatal DA loss. These findings are consistent with silent neurotoxicity, where developmental exposure to dieldrin primes the nigrostriatal striatal system to have an exacerbated response to synucleinopathy in the absence of observable changes in typical markers of nigrostriatal dysfunction and degeneration.
Collapse
Affiliation(s)
- Sierra L Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Joseph R Patterson
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Sydney A Zimmerman
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Mason R Kolanowski
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Joseph J Neubecker
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Ramsson
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
6
|
Goldstein DS, Pekker MJ, Sullivan P, Isonaka R, Sharabi Y. Modeling the Progression of Cardiac Catecholamine Deficiency in Lewy Body Diseases. J Am Heart Assoc 2022; 11:e024411. [PMID: 35621196 PMCID: PMC9238705 DOI: 10.1161/jaha.121.024411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/15/2022] [Indexed: 11/16/2022]
Abstract
Background Lewy body diseases (LBDs) feature deficiency of the sympathetic neurotransmitter norepinephrine in the left ventricular myocardium and sympathetic intra-neuronal deposition of the protein alpha-synuclein (αS). LBDs therefore are autonomic synucleinopathies. Computational modeling has revealed multiple functional abnormalities in residual myocardial sympathetic noradrenergic nerves in LBDs, including decreased norepinephrine synthesis, vesicular storage, and recycling. We report an extended model that enables predictions about the progression of LBDs and effects of genetic predispositions and treatments on that progression. Methods and Results The model combines cardiac sympathetic activation with autotoxicity mediated by the dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. We tested the model by its ability to predict longitudinal empirical data based on cardiac sympathetic neuroimaging, effects of genetic variations related to particular intra-neuronal reactions, treatment by monoamine oxidase inhibition to decrease 3,4-dihydroxyphenylacetaldehyde production, and post-mortem myocardial tissue contents of catecholamines and αS. The new model generated a triphasic decline in myocardial norepinephrine content. This pattern was confirmed by empirical data from serial cardiac 18F-dopamine positron emission tomographic scanning in patients with LBDs. The model also correctly predicted empirical data about effects of genetic variants and monoamine oxidase inhibition and about myocardial levels of catecholamines and αS. Conclusions The present computational model predicts a triphasic decline in myocardial norepinephrine content as LBDs progress. According to the model, disease-modifying interventions begun at the transition from the first to the second phase delay the onset of symptomatic disease. Computational modeling coupled with biomarkers of preclinical autonomic synucleinopathy may enable early detection and more effective treatment of LBDs.
Collapse
Affiliation(s)
- David S. Goldstein
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Mark J. Pekker
- Mathematical SciencesUniversity of Alabama at HuntsvilleHuntsvilleAL
| | - Patti Sullivan
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Risa Isonaka
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Yehonatan Sharabi
- Tel Aviv University Sackler Faculty of Medicine and Chaim Sheba Medical CenterTel HaShomerIsrael
| |
Collapse
|
7
|
Mei Y, Zhang QW, Gu Q, Liu Z, He X, Tian Y. Pillar[5]arene-Based Fluorescent Sensor Array for Biosensing of Intracellular Multi-neurotransmitters through Host-Guest Recognitions. J Am Chem Soc 2022; 144:2351-2359. [PMID: 35099950 DOI: 10.1021/jacs.1c12959] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurotransmitters are very important for neuron events and brain diseases. However, effective probes for analyzing specific neurotransmitters are currently lacking. Herein, we design and create a supramolecular fluorescent probe (CN-DFP5) by synthesizing a dual-functionalized fluorescent pillar[5]arene derivative with borate naphthalene and aldehyde coumarin recognition groups to identify large-scale neurotransmitters. The developed probe can detect seven model neurotransmitters by generating different fluorescence patterns through three types of host-guest interactions. The obtained signals are statistically processed by principal component analysis, thus the high-throughput analysis of neurotransmitters is realized under dual-channel fluorescence responses. The present probe combines the advantages of small-molecule-based probes to easily enter into living neurons and cross-reactive sensor arrays. Thus, the selective binding enables this probe to identify specific neurotransmitters in biofluids, living neurons, and tissues. High selectivity and sensitivity further demonstrate that the molecular device could extend to more applications to detect and image neurotransmitters.
Collapse
Affiliation(s)
- Yuxiao Mei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Qi-Wei Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Qingyi Gu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Zhichao Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Xiao He
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| |
Collapse
|
8
|
The Neuroprotective Role of Polydatin: Neuropharmacological Mechanisms, Molecular Targets, Therapeutic Potentials, and Clinical Perspective. Molecules 2021; 26:molecules26195985. [PMID: 34641529 PMCID: PMC8513080 DOI: 10.3390/molecules26195985] [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: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/09/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are one of the leading causes of death and disability in humans. From a mechanistic perspective, the complexity of pathophysiological mechanisms contributes to NDDs. Therefore, there is an urgency to provide novel multi-target agents towards the simultaneous modulation of dysregulated pathways against NDDs. Besides, their lack of effectiveness and associated side effects have contributed to the lack of conventional therapies as suitable therapeutic agents. Prevailing reports have introduced plant secondary metabolites as promising multi-target agents in combating NDDs. Polydatin is a natural phenolic compound, employing potential mechanisms in fighting NDDs. It is considered an auspicious phytochemical in modulating neuroinflammatory/apoptotic/autophagy/oxidative stress signaling mediators such as nuclear factor-κB (NF-κB), NF-E2–related factor 2 (Nrf2)/antioxidant response elements (ARE), matrix metalloproteinase (MMPs), interleukins (ILs), phosphoinositide 3-kinases (PI3K)/protein kinase B (Akt), and the extracellular regulated kinase (ERK)/mitogen-activated protein kinase (MAPK). Accordingly, polydatin potentially counteracts Alzheimer’s disease, cognition/memory dysfunction, Parkinson’s disease, brain/spinal cord injuries, ischemic stroke, and miscellaneous neuronal dysfunctionalities. The present study provides all of the neuroprotective mechanisms of polydatin in various NDDs. Additionally, the novel delivery systems of polydatin are provided regarding increasing its safety, solubility, bioavailability, and efficacy, as well as developing a long-lasting therapeutic concentration of polydatin in the central nervous system, possessing fewer side effects.
Collapse
|
9
|
Dagra A, Miller DR, Lin M, Gopinath A, Shaerzadeh F, Harris S, Sorrentino ZA, Støier JF, Velasco S, Azar J, Alonge AR, Lebowitz JJ, Ulm B, Bu M, Hansen CA, Urs N, Giasson BI, Khoshbouei H. α-Synuclein-induced dysregulation of neuronal activity contributes to murine dopamine neuron vulnerability. NPJ Parkinsons Dis 2021; 7:76. [PMID: 34408150 PMCID: PMC8373893 DOI: 10.1038/s41531-021-00210-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pathophysiological damages and loss of function of dopamine neurons precede their demise and contribute to the early phases of Parkinson's disease. The presence of aberrant intracellular pathological inclusions of the protein α-synuclein within ventral midbrain dopaminergic neurons is one of the cardinal features of Parkinson's disease. We employed molecular biology, electrophysiology, and live-cell imaging to investigate how excessive α-synuclein expression alters multiple characteristics of dopaminergic neuronal dynamics and dopamine transmission in cultured dopamine neurons conditionally expressing GCaMP6f. We found that overexpression of α-synuclein in mouse (male and female) dopaminergic neurons altered neuronal firing properties, calcium dynamics, dopamine release, protein expression, and morphology. Moreover, prolonged exposure to the D2 receptor agonist, quinpirole, rescues many of the alterations induced by α-synuclein overexpression. These studies demonstrate that α-synuclein dysregulation of neuronal activity contributes to the vulnerability of dopaminergic neurons and that modulation of D2 receptor activity can ameliorate the pathophysiology. These findings provide mechanistic insights into the insidious changes in dopaminergic neuronal activity and neuronal loss that characterize Parkinson's disease progression with significant therapeutic implications.
Collapse
Affiliation(s)
- Abeer Dagra
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Douglas R. Miller
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Min Lin
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adithya Gopinath
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Fatemeh Shaerzadeh
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Sharonda Harris
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Zachary A. Sorrentino
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Jonatan Fullerton Støier
- grid.5254.60000 0001 0674 042XMolecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Velasco
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Janelle Azar
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adetola R. Alonge
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Joseph J. Lebowitz
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Brittany Ulm
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Mengfei Bu
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Carissa A. Hansen
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Nikhil Urs
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Benoit I. Giasson
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Habibeh Khoshbouei
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| |
Collapse
|
10
|
Lee JH, Kim HJ, Kim JU, Yook TH, Kim KH, Lee JY, Yang G. A Novel Treatment Strategy by Natural Products in NLRP3 Inflammasome-Mediated Neuroinflammation in Alzheimer's and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22031324. [PMID: 33525754 PMCID: PMC7866084 DOI: 10.3390/ijms22031324] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are the most common neurodegenerative diseases. Many studies have demonstrated that the release of NLRP3 inflammasome-mediated proinflammatory cytokines by the excessive activation of microglia is associated with the pathogenesis of AD and PD and suggested that the NLRP3 inflammasome plays an important role in AD and PD development. In both diseases, various stimuli, such as Aβ and α-synuclein, accelerate the formation of the NLRP3 inflammasome in microglia and induce pyroptosis through the expression of interleukin (IL)-1β, caspase-1, etc., where neuroinflammation contributes to gradual progression and deterioration. However, despite intensive research, the exact function and regulation of the NLRP3 inflammasome has not yet been clearly identified. Moreover, there have not yet been any experiments of clinical use, although many studies have recently been conducted to improve treatment of inflammatory diseases using various inhibitors for NLRP3 inflammasome pathways. However, recent studies have reported that various natural products show improvement effects in the in vivo models of AD and PD through the regulation of NLRP3 inflammasome assembly. Therefore, the present review provides an overview of natural extraction studies aimed at the prevention or treatment of NLRP3 inflammasome-mediated neurological disorders. It is suggested that the discovery and development of these various natural products could be a potential strategy for NLRP3 inflammasome-mediated AD and PD treatment.
Collapse
Affiliation(s)
- Jun Ho Lee
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
| | - Hong Jun Kim
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
| | - Jong Uk Kim
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
| | - Tae Han Yook
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
| | - Kyeong Han Kim
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
| | - Joo Young Lee
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea;
| | - Gabsik Yang
- College of Korea Medicine, Woosuk University, Jeonju-si, Jeollabuk-do 54986, Korea; (J.H.L.); (H.J.K.); (J.U.K.); (T.H.Y.); (K.H.K.)
- Correspondence: ; Tel.: +82-63-290-9030
| |
Collapse
|
11
|
Mészáros Á, Molnár K, Nógrádi B, Hernádi Z, Nyúl-Tóth Á, Wilhelm I, Krizbai IA. Neurovascular Inflammaging in Health and Disease. Cells 2020; 9:cells9071614. [PMID: 32635451 PMCID: PMC7407516 DOI: 10.3390/cells9071614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022] Open
Abstract
Aging is characterized by a chronic low-grade sterile inflammation dubbed as inflammaging, which in part originates from accumulating cellular debris. These, acting as danger signals with many intrinsic factors such as cytokines, are sensed by a network of pattern recognition receptors and other cognate receptors, leading to the activation of inflammasomes. Due to the inflammasome activity-dependent increase in the levels of pro-inflammatory interleukins (IL-1β, IL-18), inflammation is initiated, resulting in tissue injury in various organs, the brain and the spinal cord included. Similarly, in age-related diseases of the central nervous system (CNS), inflammasome activation is a prominent moment, in which cells of the neurovascular unit occupy a significant position. In this review, we discuss the inflammatory changes in normal aging and summarize the current knowledge on the role of inflammasomes and contributing mechanisms in common CNS diseases, namely Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and stroke, all of which occur more frequently with aging.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Theoretical Medicine Doctoral School, University of Szeged, 6720 Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Zsófia Hernádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
| | - István A. Krizbai
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
- Correspondence: ; Tel.: +36-62-599-794
| |
Collapse
|
12
|
Zhao H, Huang S, Palanisamy S, Wang C, Rainer G, Zhang X. Alpha-Synuclein Dopaminylation Presented in Plasma of Both Healthy Subjects and Parkinson's Disease Patients. Proteomics Clin Appl 2020; 14:e1900117. [PMID: 32538547 DOI: 10.1002/prca.201900117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 05/03/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE Alpha-synuclein (α-syn) dopaminylation can lead to the death of dopaminergic neurons in the brain and is a risk factor of Parkinson's disease (PD). This study aims to examine whether such a posttranslational modification (PTM) is presented in human blood plasma. EXPERIMENTAL DESIGN In vitro reaction simulation between α-syn and dopamine (DA) is conducted to study the biochemical mechanism. Then α-syn from human blood plasma samples is detected by using immunoprecipitation-mass spectrometry (IP-MS). Lastly the levels of endogenous α-syn and α-syn dopaminylation in 88 blood plasma samples from patients with PD, major depressive disorder (MDD), and healthy control (HC) are compared. RESULTS DA modifies α-syn with the addition of dopamine-quinone (DAQ) into lysine sites of α-syn in vitro and the addition of DAQ and 3,4-dihydroxyphenylacetaldehyde (DOPAL) in plasma samples. The unmodified α-syn between the PD and HC groups showed similar levels. The levels of two peptides, one with lysine 34 (34 K) DAQ modification and the other with lysine 23 (23 K) ubiquitination, are significantly higher in PD and MDD compared with HC. CONCLUSIONS AND CLINICAL RELEVANCE Thus, α-syn dopaminylation is measurable and might be used to indicatethe presence and progression of neurological disorders.
Collapse
Affiliation(s)
- Huiyuan Zhao
- Section of Medicine, University of Fribourg, Fribourg, CH1700, Switzerland.,Division of Biological Technology, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China, 116023
| | - Shuai Huang
- Division of Biological Technology, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China, 116023
| | - Sivakumar Palanisamy
- Division of Biological Technology, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China, 116023
| | - Cui Wang
- Department of Neurology, Dalian Central Hospital, Dalian, China, 116033
| | - Gregor Rainer
- Section of Medicine, University of Fribourg, Fribourg, CH1700, Switzerland
| | - Xiaozhe Zhang
- Division of Biological Technology, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China, 116023
| |
Collapse
|
13
|
Kaehler K, Seitter H, Sandbichler AM, Tschugg B, Obermair GJ, Stefanova N, Koschak A. Assessment of the Retina of Plp-α-Syn Mice as a Model for Studying Synuclein-Dependent Diseases. Invest Ophthalmol Vis Sci 2020; 61:12. [PMID: 32503050 PMCID: PMC7415298 DOI: 10.1167/iovs.61.6.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Synucleinopathies such as multiple system atrophy (MSA) and Parkinson's disease are associated with a variety of visual symptoms. Functional and morphological retinal aberrations are therefore supposed to be valuable biomarkers for these neurodegenerative diseases. This study examined the retinal morphology and functionality resulting from human α-synuclein (α-Syn) overexpression in the transgenic Plp-α-Syn mouse model. Methods Immunohistochemistry on retinal sections and whole-mounts was performed on 8- to 11-week-old and 12-month-old Plp-α-Syn mice and C57BL/6N controls. Quantitative RT-PCR experiments were performed to study the expression of endogenous and human α-Syn and tyrosine hydroxylase (TH). We confirmed the presence of human α-Syn in the retina in western blot analyses. Multi-electrode array (MEA) analyses from light-stimulated whole-mounted retinas were used to investigate their functionality. Results Biochemical and immunohistochemical analyses showed human α-Syn in the retina of Plp-α-Syn mice. We found distinct staining in different retinal cell layers, most abundantly in rod bipolar cells of the peripheral retina. In the periphery, we also observed a trend toward a decline in the number of retinal ganglion cells. The number of TH+ neurons was unaffected in this human α-Syn overexpression model. MEA recordings showed that Plp-α-Syn retinas were functional but exhibited mild alterations in dim light conditions. Conclusions Together, these findings implicate an impairment of retinal neurons in the Plp-α-Syn mouse. The phenotype partly relates to retinal deficits reported in MSA patients. We further propose the suitability of the Plp-α-Syn retina as a biological model to study synuclein-mediated mechanisms.
Collapse
Affiliation(s)
- Kathrin Kaehler
- Institute of Pharmacy, Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Hartwig Seitter
- Institute of Pharmacy, Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| | | | - Bettina Tschugg
- Institute of Pharmacy, Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Gerald J. Obermair
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
- Physiology Division, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Institute of Pharmacy, Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Santos PI, Outeiro TF. Protein trapping leads to altered synaptic proteostasis in synucleinopathies. FEBS J 2020; 287:5294-5303. [PMID: 32400966 DOI: 10.1111/febs.15364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/01/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is associated with the accumulation of alpha-synuclein (aSyn) in intracellular inclusions known as Lewy bodies and Lewy neurites. Under physiological conditions, aSyn is found at the presynaptic terminal and exists in a dynamic equilibrium between soluble, membrane-associated and aggregated forms. Emerging evidence suggests that, under pathological conditions, aSyn begins to accumulate and acquire a toxic function at the synapse, impairing their normal function and connectivity. However, the precise molecular mechanisms linking aSyn accumulation and synaptic dysfunction are still elusive. Here, we provide an overview of our current findings and discuss the hypothesis that certain aSyn aggregates may interact with proteins with whom aSyn normally does not interact with, thereby trapping them and preventing them from performing their normal functions in the cell. We posit that such abnormal interactions start to occur during the prodromal stages of PD, eventually resulting in the overt manifestation of clinical features. Therefore, understanding the nature and behaviour of toxic aSyn species and their contribution to aSyn-mediated toxicity is crucial for the development of therapeutic strategies capable of modifying disease progression in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Patrícia I Santos
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Haque ME, Akther M, Jakaria M, Kim IS, Azam S, Choi DK. Targeting the microglial NLRP3 inflammasome and its role in Parkinson's disease. Mov Disord 2019; 35:20-33. [PMID: 31680318 DOI: 10.1002/mds.27874] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/20/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022] Open
Abstract
Excessive activation of microglia and subsequent release of proinflammatory cytokines play a crucial role in neuroinflammation and neurodegeneration in Parkinson's disease (PD). Components of the nucleotide-binding oligomerization domain and leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome complex, leucine-rich-repeat- and pyrin-domain-containing 3, caspase-1, and apoptosis-associated speck-like protein containing a CARD, are highly expressed in activated microglia in PD patient brains. Findings suggest that neurotoxins, aggregation of α-synuclein, mitochondrial reactive oxygen species, and disrupted mitophagy are the key regulators of microglial leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome activation and release of interleukin-1β and interleukin-18 caspase-1-mediated pyroptotic cell death in the substantia nigra of the brain. Although this evidence suggests the leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome may be a potential drug target for treatment of PD, the exact mechanism of how the microglia sense these stimuli and initiate leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome signaling is unknown. Here, the molecular mechanism and regulation of microglial leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome activation and its role in the pathogenesis of PD are discussed. Moreover, the potential of both endogenous and synthetic leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome modulators, long noncoding RNA, microRNA to develop novel therapeutics to treat PD is presented. Overall, we recommend that the microglial leucine-rich-repeat- and pyrin-domain-containing 3 inflammasome can be a potential target for PD treatment. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Md Ezazul Haque
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Mahbuba Akther
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Md Jakaria
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - In-Su Kim
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju, Republic of Korea
| | - Shofiul Azam
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea.,Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju, Republic of Korea
| |
Collapse
|
16
|
Masato A, Plotegher N, Boassa D, Bubacco L. Impaired dopamine metabolism in Parkinson's disease pathogenesis. Mol Neurodegener 2019; 14:35. [PMID: 31488222 PMCID: PMC6728988 DOI: 10.1186/s13024-019-0332-6] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
A full understanding of Parkinson's Disease etiopathogenesis and of the causes of the preferential vulnerability of nigrostriatal dopaminergic neurons is still an unsolved puzzle. A multiple-hit hypothesis has been proposed, which may explain the convergence of familial, environmental and idiopathic forms of the disease. Among the various determinants of the degeneration of the neurons in Substantia Nigra pars compacta, in this review we will focus on the endotoxicity associated to dopamine dyshomeostasis. In particular, we will discuss the relevance of the reactive dopamine metabolite 3,4-dihydroxyphenylacetaldehyde (DOPAL) in the catechol-induced neurotoxicity. Indeed, the synergy between the catechol and the aldehyde moieties of DOPAL exacerbates its reactivity, resulting in modification of functional protein residues, protein aggregation, oxidative stress and cell death. Interestingly, αSynuclein, whose altered proteostasis is a recurrent element in Parkinson's Disease pathology, is considered a preferential target of DOPAL modification. DOPAL triggers αSynuclein oligomerization leading to synapse physiology impairment. Several factors can be responsible for DOPAL accumulation at the pre-synaptic terminals, i.e. dopamine leakage from synaptic vesicles, increased rate of dopamine conversion to DOPAL by upregulated monoamine oxidase and decreased DOPAL degradation by aldehyde dehydrogenases. Various studies report the decreased expression and activity of aldehyde dehydrogenases in parkinsonian brains, as well as genetic variants associated to increased risk in developing the pathology. Thus, we discuss how the deregulation of these enzymes might be considered a contributing element in the pathogenesis of Parkinson's Disease or a down-stream effect. Finally, we propose that a better understanding of the impaired dopamine metabolism in Parkinson's Disease would allow a more refined patients stratification and the design of more targeted and successful therapeutic strategies.
Collapse
Affiliation(s)
- Anna Masato
- Department of Biology, University of Padova, Padova, Italy
| | | | - Daniela Boassa
- Department of Neurosciences, and National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, USA
| | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
17
|
Oliveri V. Toward the discovery and development of effective modulators of α-synuclein amyloid aggregation. Eur J Med Chem 2019; 167:10-36. [PMID: 30743095 DOI: 10.1016/j.ejmech.2019.01.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
A host of human diseases, including Parkinson's disease and Dementia with Lewy bodies, are suspected to be directly linked to protein aggregation. Amyloid protein aggregates and oligomeric intermediates of α-synuclein are observed in synucleinopathies and considered to be mediators of cellular toxicity. Hence, α-synuclein has seen as one of the leading and most compelling targets and is receiving a great deal of attention from researchers. Nevertheless, there is no neuroprotective approach directed toward Parkinson's disease or other synucleinopathies so far. In this review, we summarize the available data concerning inhibitors of α-synuclein aggregation and their advancing towards clinical use. The compounds are grouped according to their chemical structures, providing respective insights into their mechanism of action, pharmacology, and pharmacokinetics. Overall, shared structure-activity elements are emerging, as well as specific binding modes related to the ability of the modulators to establish hydrophobic and hydrogen bonds interactions with the protein. Some molecules with encouraging in vivo data support the possibility of translation to the clinic.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, viale A. Doria 6, 95125, Catania, Italy.
| |
Collapse
|
18
|
Lang AE, Espay AJ. Disease Modification in Parkinson's Disease: Current Approaches, Challenges, and Future Considerations. Mov Disord 2018; 33:660-677. [DOI: 10.1002/mds.27360] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/04/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Anthony E. Lang
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Division of Neurology; University of Toronto; Toronto Ontario Canada
| | - Alberto J. Espay
- UC Gardner Neuroscience Institute and Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology; University of Cincinnati; Cincinnati Ohio USA
| |
Collapse
|
19
|
Abstract
Parkinson's disease (PD) is characterized by intracellular inclusions of aggregated and misfolded α-Synuclein (α-Syn), and the loss of dopaminergic (DA) neurons in the brain. The resulting motor abnormalities mark the progression of PD, while non-motor symptoms can already be identified during early, prodromal stages of disease. Recent studies provide evidence that during this early prodromal phase, synaptic and axonal abnormalities occur before the degenerative loss of neuronal cell bodies. These early phenotypes can be attributed to synaptic accumulation of toxic α-Syn. Under physiological conditions, α-Syn functions in its native conformation as a soluble monomer. However, PD patient brains are characterized by intracellular inclusions of insoluble fibrils. Yet, oligomers and protofibrils of α-Syn have been identified to be the most toxic species, with their accumulation at presynaptic terminals affecting several steps of neurotransmitter release. First, high levels of α-Syn alter the size of synaptic vesicle pools and impair their trafficking. Second, α-Syn overexpression can either misregulate or redistribute proteins of the presynaptic SNARE complex. This leads to deficient tethering, docking, priming and fusion of synaptic vesicles at the active zone (AZ). Third, α-Syn inclusions are found within the presynaptic AZ, accompanied by a decrease in AZ protein levels. Furthermore, α-Syn overexpression reduces the endocytic retrieval of synaptic vesicle membranes during vesicle recycling. These presynaptic alterations mediated by accumulation of α-Syn, together impair neurotransmitter exocytosis and neuronal communication. Although α-Syn is expressed throughout the brain and enriched at presynaptic terminals, DA neurons are the most vulnerable in PD, likely because α-Syn directly regulates dopamine levels. Indeed, evidence suggests that α-Syn is a negative modulator of dopamine by inhibiting enzymes responsible for its synthesis. In addition, α-Syn is able to interact with and reduce the activity of VMAT2 and DAT. The resulting dysregulation of dopamine levels directly contributes to the formation of toxic α-Syn oligomers. Together these data suggest a vicious cycle of accumulating α-Syn and deregulated dopamine that triggers synaptic dysfunction and impaired neuronal communication, ultimately causing synaptopathy and progressive neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Jessika C Bridi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Frank Hirth
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| |
Collapse
|