1
|
Combes BF, Kalva SK, Benveniste PL, Tournant A, Law MH, Newton J, Krüger M, Weber RZ, Dias I, Noain D, Dean-Ben XL, Konietzko U, Baumann CR, Gillberg PG, Hock C, Nitsch RM, Cohen-Adad J, Razansky D, Ni R. Spiral volumetric optoacoustic tomography of reduced oxygen saturation in the spinal cord of M83 mouse model of Parkinson's disease. Eur J Nucl Med Mol Imaging 2025; 52:427-443. [PMID: 39382580 PMCID: PMC11732882 DOI: 10.1007/s00259-024-06938-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE Metabolism and bioenergetics in the central nervous system play important roles in the pathophysiology of Parkinson's disease (PD). Here, we employed a multimodal imaging approach to assess oxygenation changes in the spinal cord of the transgenic M83 murine model of PD overexpressing the mutated A53T alpha-synuclein form in comparison with non-transgenic littermates. METHODS In vivo spiral volumetric optoacoustic tomography (SVOT) was performed to assess oxygen saturation (sO2) in the spinal cords of M83 mice and non-transgenic littermates. Ex vivo high-field T1-weighted (T1w) magnetic resonance imaging (MRI) at 9.4T was used to assess volumetric alterations in the spinal cord. 3D SVOT analysis and deep learning-based automatic segmentation of T1w MRI data for the mouse spinal cord were developed for quantification. Immunostaining for phosphorylated alpha-synuclein (pS129 α-syn), as well as vascular organization (CD31 and GLUT1), was performed after MRI scan. RESULTS In vivo SVOT imaging revealed a lower sO2SVOT in the spinal cord of M83 mice compared to non-transgenic littermates at sub-100 μm spatial resolution. Ex vivo MRI-assisted by in-house developed deep learning-based automatic segmentation (validated by manual analysis) revealed no volumetric atrophy in the spinal cord of M83 mice compared to non-transgenic littermates at 50 μm spatial resolution. The vascular network was not impaired in the spinal cord of M83 mice in the presence of pS129 α-syn accumulation. CONCLUSION We developed tools for deep-learning-based analysis for the segmentation of mouse spinal cord structural MRI data, and volumetric analysis of sO2SVOT data. We demonstrated non-invasive high-resolution imaging of reduced sO2SVOT in the absence of volumetric structural changes in the spinal cord of PD M83 mouse model.
Collapse
Affiliation(s)
- Benjamin F Combes
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Sandeep Kumar Kalva
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Pierre-Louis Benveniste
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| | - Agathe Tournant
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Man Hoi Law
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Joshua Newton
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Maik Krüger
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Inês Dias
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| | - Xose Luis Dean-Ben
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| | - Per-Göran Gillberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
2
|
Burré J, Edwards RH, Halliday G, Lang AE, Lashuel HA, Melki R, Murayama S, Outeiro TF, Papa SM, Stefanis L, Woerman AL, Surmeier DJ, Kalia LV, Takahashi R. Research Priorities on the Role of α-Synuclein in Parkinson's Disease Pathogenesis. Mov Disord 2024; 39:1663-1678. [PMID: 38946200 DOI: 10.1002/mds.29897] [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: 01/05/2024] [Revised: 05/16/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Various forms of Parkinson's disease, including its common sporadic form, are characterized by prominent α-synuclein (αSyn) aggregation in affected brain regions. However, the role of αSyn in the pathogenesis and evolution of the disease remains unclear, despite vast research efforts of more than a quarter century. A better understanding of the role of αSyn, either primary or secondary, is critical for developing disease-modifying therapies. Previous attempts to hone this research have been challenged by experimental limitations, but recent technological advances may facilitate progress. The Scientific Issues Committee of the International Parkinson and Movement Disorder Society (MDS) charged a panel of experts in the field to discuss current scientific priorities and identify research strategies with potential for a breakthrough. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jacqueline Burré
- Appel Institute for Alzheimer's Disease Research and Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Robert H Edwards
- Department of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, California, USA
| | - Glenda Halliday
- Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hilal A Lashuel
- Laboratory of Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses, France
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- The Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Stella M Papa
- Department of Neurology, School of Medicine, and Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Leonidas Stefanis
- First Department of Neurology, Eginitio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Amanda L Woerman
- Department of Biology, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, Colorado, USA
| | - Dalton James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Lorraine V Kalia
- Edmond J. Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Akyazı O, Korkmaz D, Cevher SC. Experimental Parkinson models and green chemistry approach. Behav Brain Res 2024; 471:115092. [PMID: 38844056 DOI: 10.1016/j.bbr.2024.115092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Parkinson's is the most common neurodegenerative disease after Alzheimer's. Motor findings in Parkinson's occur as a result of the degeneration of dopaminergic neurons starting in the substantia nigra pars compacta and ending in the putamen and caudate nucleus. Loss of neurons and the formation of inclusions called Lewy bodies in existing neurons are characteristic histopathological findings of Parkinson's. The disease primarily impairs the functional capacity of the person with cardinal findings such as tremor, bradykinesia, etc., as a result of the loss of dopaminergic neurons in the substantia nigra. Experimental animal models of Parkinson's have been used extensively in recent years to investigate the pathology of this disease. These models are generally based on systemic or local(intracerebral) administration of neurotoxins, which can replicate many features of Parkinson's mammals. The development of transgenic models in recent years has allowed us to learn more about the modeling of Parkinson's. Applying animal modeling, which shows the most human-like effects in studies, is extremely important. It has been demonstrated that oxidative stress increases in many neurodegenerative diseases such as Parkinson's and various age-related degenerative diseases in humans and that neurons are sensitive to it. In cases where oxidative stress increases and antioxidant systems are inadequate, natural molecules such as flavonoids and polyphenols can be used as a new antioxidant treatment to reduce neuronal reactive oxygen species and improve the neurodegenerative process. Therefore, in this article, we examined experimental animal modeling in Parkinson's disease and the effect of green chemistry approaches on Parkinson's disease.
Collapse
Affiliation(s)
- Ozge Akyazı
- Gazi University, Institute of Science, Department of Biology, Ankara 06500, Turkey.
| | - Dılara Korkmaz
- Gazi University, Institute of Science, Department of Biology, Ankara 06500, Turkey
| | - Sule Coskun Cevher
- Gazi University, Faculty of Science, Department of Biology, Ankara 06500, Turkey
| |
Collapse
|
4
|
Liquid-liquid Phase Separation of α-Synuclein: A New Mechanistic Insight for α-Synuclein Aggregation Associated with Parkinson's Disease Pathogenesis. J Mol Biol 2023; 435:167713. [PMID: 35787838 DOI: 10.1016/j.jmb.2022.167713] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Aberrant aggregation of the misfolded presynaptic protein, α-Synuclein (α-Syn) into Lewy body (LB) and Lewy neuritis (LN) is a major pathological hallmark of Parkinson's disease (PD) and other synucleinopathies. Numerous studies have suggested that prefibrillar and fibrillar species of the misfolded α-Syn aggregates are responsible for cell death in PD pathogenesis. However, the precise molecular events during α-Syn aggregation, especially in the early stages, remain elusive. Emerging evidence has demonstrated that liquid-liquid phase separation (LLPS) of α-Syn occurs in the nucleation step of α-Syn aggregation, which offers an alternate non-canonical aggregation pathway in the crowded microenvironment. The liquid-like α-Syn droplets gradually undergo an irreversible liquid-to-solid phase transition into amyloid-like hydrogel entrapping oligomers and fibrils. This new mechanism of α-Syn LLPS and gel formation might represent the molecular basis of cellular toxicity associated with PD. This review aims to demonstrate the recent development of α-Syn LLPS, the underlying mechanism along with the microscopic events of aberrant phase transition. This review further discusses how several intrinsic and extrinsic factors regulate the thermodynamics and kinetics of α-Syn LLPS and co-LLPS with other proteins, which might explain the pathophysiology of α-Syn in various neurodegenerative diseases.
Collapse
|
5
|
See WZC, Naidu R, Tang KS. Cellular and Molecular Events Leading to Paraquat-Induced Apoptosis: Mechanistic Insights into Parkinson’s Disease Pathophysiology. Mol Neurobiol 2022; 59:3353-3369. [PMID: 35306641 PMCID: PMC9148284 DOI: 10.1007/s12035-022-02799-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 03/09/2022] [Indexed: 12/17/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the cardinal features of tremor, bradykinesia, rigidity, and postural instability, in addition to other non-motor symptoms. Pathologically, PD is attributed to the loss of dopaminergic neurons in the substantia nigra pars compacta, with the hallmark of the presence of intracellular protein aggregates of α-synuclein in the form of Lewy bodies. The pathogenesis of PD is still yet to be fully elucidated due to the multifactorial nature of the disease. However, a myriad of studies has indicated several intracellular events in triggering apoptotic neuronal cell death in PD. These include oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, alteration in dopamine catabolism, inactivation of tyrosine hydroxylase, and decreased levels of neurotrophic factors. Laboratory studies using the herbicide paraquat in different in vitro and in vivo models have demonstrated the induction of many PD pathological features. The selective neurotoxicity induced by paraquat has brought a new dawn in our perspectives about the pathophysiology of PD. Epidemiological data have suggested an increased risk of developing PD in the human population exposed to paraquat for a long term. This model has opened new frontiers in the quest for new therapeutic targets for PD. The purpose of this review is to synthesize the relationship between the exposure of paraquat and the pathogenesis of PD in in vitro and in vivo models.
Collapse
Affiliation(s)
- Wesley Zhi Chung See
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Kim San Tang
- School of Pharmacy, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
6
|
Mani S, Sevanan M, Krishnamoorthy A, Sekar S. A systematic review of molecular approaches that link mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurol Sci 2021; 42:4459-4469. [PMID: 34480241 DOI: 10.1007/s10072-021-05551-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/07/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder that affects 1% of the population worldwide. Etiology of PD is likely to be multi-factorial such as protein misfolding, mitochondrial dysfunction, oxidative stress, and neuroinflammation that contributes to the pathology of Parkinson's disease (PD), numerous studies have shown that mitochondrial dysfunction may play a key role in the dopaminergic neuronal loss. In multiple ways, the two most important are the activation of neuroinflammation and mitochondrial dysfunction, while mitochondrial dysfunction could cause neuroinflammation and vice versa. Thus, the mitochondrial proteins are the highly promising target for the development of PD. However, the limited amount of dopaminergic neurons prevented the detailed investigation of Parkinson's disease with regard to mitochondrial dysfunction. Both genetic and environmental factors are also associated with mitochondrial dysfunction and PD pathogenesis. The induction of PD by neurotoxins that inhibit mitochondrial complex I provide direct evidence linking mitochondrial dysfunction to PD. A decrease of mitochondrial complex I activity is observed in PD brain and in neurotoxin- or genetic factor-induced in vitro and in vivo models. Moreover, PINK1, Parkin, DJ-1 and LRRK2 mitochondrial PD gene products have important roles in mitophagy, a cellular process that clear damaged mitochondria. This review paper would discuss the evidence for the mitochondrial dysfunction and neuroinflammation in PD.
Collapse
Affiliation(s)
- Sugumar Mani
- Research and Development Centre, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Murugan Sevanan
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Karunya Nagar, Coimbatore, Tamil Nadu, 641114, India.
| | | | - Sathiya Sekar
- Department of Biotechnology, Dr.M.G.R Educational Research Institute, Chennai, India
| |
Collapse
|
7
|
Qiu X, Wang Q, Hou L, Zhang C, Wang Q, Zhao X. Inhibition of NLRP3 inflammasome by glibenclamide attenuated dopaminergic neurodegeneration and motor deficits in paraquat and maneb-induced mouse Parkinson's disease model. Toxicol Lett 2021; 349:1-11. [PMID: 34052309 DOI: 10.1016/j.toxlet.2021.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/26/2021] [Accepted: 05/25/2021] [Indexed: 01/24/2023]
Abstract
Pesticides exposure can lead to damage of dopaminergic neurons, which are associated with increased risk of Parkinson's disease (PD). However, the etiology of PD remains poorly understood and no therapeutic strategy is available. Previous studies suggested the involvement of NLRP3 inflammasome in the onset of PD. This study was designed to investigate whether glibenclamide, an inhibitor of NLRP3 inflammasome, could offer a reliable protective strategy for PD in a mouse PD model induced by paraquat and maneb. We found that glibenclamide exerted potent neuroprotection against paraquat and maneb-induced upregulation of α-synuclein, dopaminergic neurodegeneration and motor impairment in brain of mice. Mechanistically, glibenclamide treatment blocked NLRP3 inflammasome activation evidenced by reduced expressions of NLRP3, activated caspase-1 and mature interleukin-1β in glibenclamide co-treated mice compared with those in paraquat and maneb group mice. Furthermore, glibenclamide treatment mitigated paraquat and maneb-induced microglial M1 proinflammatory response and nuclear factor-κB activation in mice. Finally, the increased superoxide production, lipid peroxidation, protein levels of NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) induced by paraquat and maneb were all attenuated by glibenclamide. Overall, our findings demonstrated that glibenclamide protected dopaminergic neurons in a mouse PD model induced by combined exposures of paraquat and maneb through suppression of NLRP3 inflammasome activation, microglial M1 polarization and oxidative stress.
Collapse
Affiliation(s)
- Xiaofei Qiu
- Qingdao Municipal Center for Disease Control & Prevention, Qingdao Institute of Preventive Medicine, Qingdao, 266033, China; School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qinghui Wang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Liyan Hou
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Cuili Zhang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, 116044, China.
| | - Xiulan Zhao
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
8
|
|
9
|
Singh A, Dawson TM, Kulkarni S. Neurodegenerative disorders and gut-brain interactions. J Clin Invest 2021; 131:e143775. [PMID: 34196307 DOI: 10.1172/jci143775] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders (NDs) affect essential functions not only in the CNS, but also cause persistent gut dysfunctions, suggesting that they have an impact on both CNS and gut-innervating neurons. Although the CNS biology of NDs continues to be well studied, how gut-innervating neurons, including those that connect the gut to the brain, are affected by or involved in the etiology of these debilitating and progressive disorders has been understudied. Studies in recent years have shown how CNS and gut biology, aided by the gut-brain connecting neurons, modulate each other's functions. These studies underscore the importance of exploring the gut-innervating and gut-brain connecting neurons of the CNS and gut function in health, as well as the etiology and progression of dysfunction in NDs. In this Review, we discuss our current understanding of how the various gut-innervating neurons and gut physiology are involved in the etiology of NDs, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, to cause progressive CNS and persistent gut dysfunction.
Collapse
Affiliation(s)
- Alpana Singh
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering.,Department of Neurology.,Solomon H. Snyder Department of Neuroscience, and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA
| | - Subhash Kulkarni
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| |
Collapse
|
10
|
Dwyer Z, Rudyk C, Farmer K, Beauchamp S, Shail P, Derksen A, Fortin T, Ventura K, Torres C, Ayoub K, Hayley S. Characterizing the protracted neurobiological and neuroanatomical effects of paraquat in a murine model of Parkinson's disease. Neurobiol Aging 2021; 100:11-21. [PMID: 33450723 DOI: 10.1016/j.neurobiolaging.2020.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/16/2020] [Accepted: 11/19/2020] [Indexed: 01/18/2023]
Abstract
The primary motor symptoms of Parkinson's disease (PD) result from the degeneration of dopamine-producing neurons of the substantia nigra pars compacta (SNc), and often, the loss is asymmetrical, resulting in unilateral tremor presentation. Notably, age is the primary risk factor for PD, and it is likely that the disease ultimately stems from the impact of environmental factors, which interact with the aging process. Recent research has focused on the role of microglia and pro-oxidative responses in dopaminergic neuronal death. In this study, we sought to examine the neurodegenerative, inflammatory, and stress effects of exposure to the etiologically relevant pesticide, paraquat, over time (up to 6 months after injections). We also were interested in whether a high-resolution, 7-Tesla animal magnetic resonance imaging would be sensitive enough to detect the degenerative impact of paraquat. We found that paraquat induced a loss of dopaminergic SNc neurons and activation of microglia that surprisingly did not change over 6 months after the last injection. A long-lasting reduction was evident for body weight, and alterations in organ (lung and heart) weight were evident, which reflect the peripheral impact of the toxicant. The microglial proinflammatory actin-remodeling factor, WAVE2, along with the inflammatory transcription factor, nuclear factor kappa B were also elevated within the brain. Remarkably, the stress hormone, corticosterone, was still significantly elevated 1 month after paraquat, whereas the inflammasome factor, caspase-1, and antigen presentation factor, MFG-E8, both displayed delayed rises after the 6-month time. Using high-resolution magnetic resonance imaging, we detected no striatal changes but modest hemispheric differences in the SNc and time-dependent volumetric enlargement of the ventricles in paraquat-treated mice. These data suggest that paraquat induces long-term nigrostriatal pathology (possibly asymmetric) and inflammatory changes and stress and trophic/apoptotic effects that appear to either increase with the passage of time or are evident for at least 1 month. In brief, paraquat may be a useful nonspecific means to model widespread stress and inflammatory changes related to PD or age-related disease in general, but not the progressive nature of such diseases.
Collapse
Affiliation(s)
- Zach Dwyer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Chris Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Kyle Farmer
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sheryl Beauchamp
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Pragya Shail
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Alexa Derksen
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Teresa Fortin
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Katelyn Ventura
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Carlos Torres
- Department of Radiology, University of Ottawa, Ottawa, Ontario, Canada; Department of Medical Imaging, The Ottawa Hospital, Ottawa, Ontario, Canada; Ottawa Hospital Research (OHRI), Ottawa, Ontario, Canada
| | - Kiara Ayoub
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
11
|
El-Gamal M, Salama M, Collins-Praino LE, Baetu I, Fathalla AM, Soliman AM, Mohamed W, Moustafa AA. Neurotoxin-Induced Rodent Models of Parkinson's Disease: Benefits and Drawbacks. Neurotox Res 2021; 39:897-923. [PMID: 33765237 DOI: 10.1007/s12640-021-00356-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by cardinal motor impairments, including akinesia and tremor, as well as by a host of non-motor symptoms, including both autonomic and cognitive dysfunction. PD is associated with a death of nigral dopaminergic neurons, as well as the pathological spread of Lewy bodies, consisting predominantly of the misfolded protein alpha-synuclein. To date, only symptomatic treatments, such as levodopa, are available, and trials aiming to cure the disease, or at least halt its progression, have not been successful. Wong et al. (2019) suggested that the lack of effective therapy against neurodegeneration in PD might be attributed to the fact that the molecular mechanisms standing behind the dopaminergic neuronal vulnerability are still a major scientific challenge. Understanding these molecular mechanisms is critical for developing effective therapy. Thirty-five years ago, Calne and William Langston (1983) raised the question of whether biological or environmental factors precipitate the development of PD. In spite of great advances in technology and medicine, this question still lacks a clear answer. Only 5-15% of PD cases are attributed to a genetic mutation, with the majority of cases classified as idiopathic, which could be linked to exposure to environmental contaminants. Rodent models play a crucial role in understanding the risk factors and pathogenesis of PD. Additionally, well-validated rodent models are critical for driving the preclinical development of clinically translatable treatment options. In this review, we discuss the mechanisms, similarities and differences, as well as advantages and limitations of different neurotoxin-induced rat models of PD. In the second part of this review, we will discuss the potential future of neurotoxin-induced models of PD. Finally, we will briefly demonstrate the crucial role of gene-environment interactions in PD and discuss fusion or dual PD models. We argue that these models have the potential to significantly further our understanding of PD.
Collapse
Affiliation(s)
- Mohamed El-Gamal
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohamed Salama
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), Dublin, Ireland
| | | | | | - Ahmed M Fathalla
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amira M Soliman
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wael Mohamed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Mansoura, Egypt.,Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University, Kuantan, Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and Marcs Institute for Brain and Behaviour, Western Sydney University, Sydney, NSW, Australia.,Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
12
|
Dwyer Z, Chaiquin M, Landrigan J, Ayoub K, Shail P, Rocha J, Childers CL, Storey KB, Philpott DJ, Sun H, Hayley S. The impact of dextran sodium sulphate and probiotic pre-treatment in a murine model of Parkinson's disease. J Neuroinflammation 2021; 18:20. [PMID: 33422110 PMCID: PMC7796536 DOI: 10.1186/s12974-020-02062-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recent work has established that Parkinson's disease (PD) patients have an altered gut microbiome, along with signs of intestinal inflammation. This could help explain the high degree of gastric disturbances in PD patients, as well as potentially be linked to the migration of peripheral inflammatory factors into the brain. To our knowledge, this is the first study to examine microbiome alteration prior to the induction of a PD murine model. METHODS We presently assessed whether pre-treatment with the probiotic, VSL #3, or the inflammatory inducer, dextran sodium sulphate (DSS), would influence the PD-like pathology provoked by a dual hit toxin model using lipopolysaccharide (LPS) and paraquat exposure. RESULTS While VSL #3 has been reported to have anti-inflammatory effects, DSS is often used as a model of colitis because of the gut inflammation and the breach of the intestinal barrier that it induces. We found that VSL#3 did not have any significant effects (beyond a blunting of LPS paraquat-induced weight loss). However, the DSS treatment caused marked changes in the gut microbiome and was also associated with augmented behavioral and inflammatory outcomes. In fact, DSS markedly increased taxa belonging to the Bacteroidaceae and Porphyromonadaceae families but reduced those from Rikencellaceae and S24-7, as well as provoking colonic pro-inflammatory cytokine expression, consistent with an inflamed gut. The DSS also increased the impact of LPS plus paraquat upon microglial morphology, along with circulating lipocalin-2 (neutrophil marker) and IL-6. Yet, neither DSS nor VSL#3 influenced the loss of substantia nigra dopamine neurons or the astrocytic and cytoskeleton remodeling protein changes that were provoked by the LPS followed by paraquat treatment. CONCLUSIONS These data suggest that disruption of the intestinal integrity and the associated microbiome can interact with systemic inflammatory events to promote widespread brain-gut changes that could be relevant for PD and at the very least, suggestive of novel neuro-immune communication.
Collapse
Affiliation(s)
- Zach Dwyer
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Melany Chaiquin
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Jeffrey Landrigan
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Kiara Ayoub
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Pragya Shail
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Julianna Rocha
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Christie L Childers
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Hongyu Sun
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada.
| |
Collapse
|
13
|
Khan Z, Ali SA. A preliminary study assessing the effect of isocyanate in neuroblastoma brain cells in vitro. Acta Neurobiol Exp (Wars) 2021. [DOI: 10.21307/ane-2021-018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
14
|
Rai SN, Singh P. Advancement in the modelling and therapeutics of Parkinson's disease. J Chem Neuroanat 2020; 104:101752. [PMID: 31996329 DOI: 10.1016/j.jchemneu.2020.101752] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/19/2020] [Accepted: 01/20/2020] [Indexed: 02/08/2023]
Abstract
Since the discovery of L-dopa in the middle of the 20th century (1960s), there is not any neuroprotective therapy available although significant development has been made in the treatment of symptomatic Parkinson's disease (PD). Neurological disorders like PD can be modelled in animals so as to recapitulates most of the symptoms seen in PD patients. In aging population, PD is the second most common neurodegenerative disease after Alzheimer's disease, even though significant outcomes have been achieved in PD research yet it still is a mystery to solve the treatments for PD. In the last two decades, PD models have provided enhanced precision into the understanding of the process of PD disease, its etiology, pathology, and molecular mechanisms behind it. Furthermore, at the same time as cellular models have helped to recognize specific events, animal models, both toxic and genetic, have replicated almost all of the hallmarks of PD and are very helpful for testing and finding new strategies for neuroprotection. Recently, in both classical and newer models, major advances have been done in the modelling of supplementary PD features have come into the light. In this review, we have try to provide an updated summary of the characteristics of these models related to in vitro and in vivo models, animal models for PD, stem cell model for PD, newer 3D model as well as the strengths and limitations of these most popular PD models.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| | - Payal Singh
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
15
|
Selective vulnerability in α-synucleinopathies. Acta Neuropathol 2019; 138:681-704. [PMID: 31006067 PMCID: PMC6800835 DOI: 10.1007/s00401-019-02010-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/13/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy are neurodegenerative disorders resulting in progressive motor/cognitive deficits among other symptoms. They are characterised by stereotypical brain cell loss accompanied by the formation of proteinaceous aggregations of the protein α-synuclein (α-syn), being, therefore, termed α-synucleinopathies. Although the presence of α-syn inclusions is a common hallmark of these disorders, the exact nature of the deposited protein is specific to each disease. Different neuroanatomical regions and cellular populations manifest a differential vulnerability to the appearance of protein deposits, cell dysfunction, and cell death, leading to phenotypic diversity. The present review describes the multiple factors that contribute to the selective vulnerability in α-synucleinopathies. We explore the intrinsic cellular properties in the affected regions, including the physiological and pathophysiological roles of endogenous α-syn, the metabolic and genetic build-up of the cells and their connectivity. These factors converge with the variability of the α-syn conformational strains and their spreading capacity to dictate the phenotypic diversity and regional vulnerability of each disease. Finally, we describe the exogenous and environmental factors that potentially contribute by igniting and modulating the differential pathology in α-synucleinopathies. In conclusion, we think that it is the confluence of this disruption of the cellular metabolic state and α-syn structural equilibrium through the anatomical connectivity which appears to initiate cascades of pathological processes triggered by genetic, environmental, or stochastic events that result in the "death by a thousand cuts" profile of α-synucleinopathies.
Collapse
|
16
|
Bullich C, Keshavarzian A, Garssen J, Kraneveld A, Perez-Pardo P. Gut Vibes in Parkinson's Disease: The Microbiota-Gut-Brain Axis. Mov Disord Clin Pract 2019; 6:639-651. [PMID: 31745471 DOI: 10.1002/mdc3.12840] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/08/2019] [Accepted: 08/20/2019] [Indexed: 12/17/2022] Open
Abstract
Background The complexity of the pathogenic mechanisms underlying neurodegenerative disorders such as Parkinson's disease (PD) is attributable to multifactorial changes occurring at a molecular level, influenced by genetics and environmental interactions. However, what causes the main hallmarks of PD is not well understood. Recent data increasingly suggest that imbalances in the gut microbiome composition might trigger and/or exacerbate the progression of PD. Objective The present review aims to (1) report emerging literature showing changes in microbiota composition of PD patients compared to healthy individuals and (2) discuss how these changes may initiate and/or perpetuate PD pathology. Methods We analyzed 13 studies published from 2015 and included in this review. Altered microbial taxa were compiled in a detailed table summarizing bacterial changes in fecal/mucosal samples. The methodology was systematically reviewed across the articles and was also included in a table to facilitate comparisons between studies. Results Multiple studies found a reduction in short-chain fatty-acid-producing bacteria that can rescue neuronal damage through epigenetic mechanisms. Overall, the studies showed that changes in the gut microbiota composition might influence colonic inflammation, gut permeability, and α-synuclein aggregation, contributing to the neurogenerative process. Conclusion Further studies with larger cohorts and high-resolution sequencing methods are required to better define gut microbiota changes in PD. Furthermore, additional longitudinal studies are required to determine the causal link between these changes and PD pathogenesis as well as to study the potential of the intestinal microbiota as a biomarker.
Collapse
Affiliation(s)
- Clara Bullich
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands
| | - Ali Keshavarzian
- Department of Medicine, Division of Allergy-Immunology Rush University Medical Center Chicago Illinois USA
| | - Johan Garssen
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands.,Nutricia Reasearch Utrecht The Netherlands
| | - Aletta Kraneveld
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands.,Institute for Risk Assessment Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands
| |
Collapse
|
17
|
Stanojlovic M, Pallais JP, Kotz CM. Chemogenetic Modulation of Orexin Neurons Reverses Changes in Anxiety and Locomotor Activity in the A53T Mouse Model of Parkinson's Disease. Front Neurosci 2019; 13:702. [PMID: 31417337 PMCID: PMC6682689 DOI: 10.3389/fnins.2019.00702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/21/2019] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD symptomology is recognized as heterogeneous and in addition to motor function decline includes cognitive, mood, sleep, and metabolic disorders. Previous studies showed early reductions in anxiety and locomotion in the A53T mice model of PD. Since inflammation and astrogliosis are an integral part of PD pathology and impair proper neuronal function, we were keen to investigate if behavioral changes in A53T mice are accompanied by increased inflammation and astrogliosis in the hippocampus (Hipp) and motor cortex (mCtx) brain regions involved in the regulation of anxiety and locomotion, respectively. To test this, we used 3-, 5-, and 7-month-old A53T mice to examine anxiety-like behavior, locomotion, and expression of inflammation and astrogliosis markers in the Hipp and mCtx. Further, we examined the presence of alpha-synuclein accumulation in orexin neurons and orexin neuronal loss. The data show early reductions in anxiety-like behavior as well as increased locomotor activity, which was accompanied by inflammation and astrogliosis in the Hipp and mCtx. Due to the persistence of the orexin neuron population in A53T mice and the involvement of orexin in anxiety and locomotor regulation, we hypothesized that chemogenetic modulation of orexin neurons would reverse the observed reductions in anxiety-like behavior and the increases in locomotor activity in these animals. We showed that chemogenetic activation of orexin neurons in A53T mice restores anxiety-like behavior back to control levels without affecting locomotor activity, whereas the inhibition of orexin neurons reverses the elevated locomotor activity without any effects on anxiety-like behavior. This study exemplifies the complex role of orexin neurons in this model of PD and demonstrates the novel finding that changes in locomotor and anxiety-like behavior are accompanied by inflammation and astrogliosis. Together, these data suggest that the orexin system may play a significant role in early and late stages of PD.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States.,Minneapolis VA Health Care System, Geriatric Research, Education and Clinical Center, Minneapolis, MN, United States
| |
Collapse
|
18
|
Seleem AA. Teratogenicity and neurotoxicity effects induced by methomyl insecticide on the developmental stages of Bufo arabicus. Neurotoxicol Teratol 2019; 72:1-9. [DOI: 10.1016/j.ntt.2018.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/22/2018] [Accepted: 12/30/2018] [Indexed: 01/01/2023]
|
19
|
Cao F, Souders Ii CL, Perez-Rodriguez V, Martyniuk CJ. Elucidating Conserved Transcriptional Networks Underlying Pesticide Exposure and Parkinson's Disease: A Focus on Chemicals of Epidemiological Relevance. Front Genet 2019; 9:701. [PMID: 30740124 PMCID: PMC6355689 DOI: 10.3389/fgene.2018.00701] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022] Open
Abstract
While a number of genetic mutations are associated with Parkinson's disease (PD), it is also widely acknowledged that the environment plays a significant role in the etiology of neurodegenerative diseases. Epidemiological evidence suggests that occupational exposure to pesticides (e.g., dieldrin, paraquat, rotenone, maneb, and ziram) is associated with a higher risk of developing PD in susceptible populations. Within dopaminergic neurons, environmental chemicals can have an array of adverse effects resulting in cell death, such as aberrant redox cycling and oxidative damage, mitochondrial dysfunction, unfolded protein response, ubiquitin-proteome system dysfunction, neuroinflammation, and metabolic disruption. More recently, our understanding of how pesticides affect cells of the central nervous system has been strengthened by computational biology. New insight has been gained about transcriptional and proteomic networks, and the metabolic pathways perturbed by pesticides. These networks and cell signaling pathways constitute potential therapeutic targets for intervention to slow or mitigate neurodegenerative diseases. Here we review the epidemiological evidence that supports a role for specific pesticides in the etiology of PD and identify molecular profiles amongst these pesticides that may contribute to the disease. Using the Comparative Toxicogenomics Database, these transcripts were compared to those regulated by the PD-associated neurotoxicant MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). While many transcripts are already established as those related to PD (alpha-synuclein, caspases, leucine rich repeat kinase 2, and parkin2), lesser studied targets have emerged as “pesticide/PD-associated transcripts” [e.g., phosphatidylinositol glycan anchor biosynthesis class C (Pigc), allograft inflammatory factor 1 (Aif1), TIMP metallopeptidase inhibitor 3, and DNA damage inducible transcript 4]. We also compared pesticide-regulated genes to a recent meta-analysis of genome-wide association studies in PD which revealed new genetic mutant alleles; the pesticides under review regulated the expression of many of these genes (e.g., ELOVL fatty acid elongase 7, ATPase H+ transporting V0 subunit a1, and bridging integrator 3). The significance is that these proteins may contribute to pesticide-related increases in PD risk. This review collates information on transcriptome responses to PD-associated pesticides to develop a mechanistic framework for quantifying PD risk with exposures.
Collapse
Affiliation(s)
- Fangjie Cao
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, College of Veterinary Medicine, University of Florida Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL, United States
| | - Christopher L Souders Ii
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, College of Veterinary Medicine, University of Florida Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL, United States
| | - Veronica Perez-Rodriguez
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, College of Veterinary Medicine, University of Florida Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL, United States
| | - Christopher J Martyniuk
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, College of Veterinary Medicine, University of Florida Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
20
|
Watts ME, Pocock R, Claudianos C. Brain Energy and Oxygen Metabolism: Emerging Role in Normal Function and Disease. Front Mol Neurosci 2018; 11:216. [PMID: 29988368 PMCID: PMC6023993 DOI: 10.3389/fnmol.2018.00216] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/01/2018] [Indexed: 01/09/2023] Open
Abstract
Dynamic metabolic changes occurring in neurons are critically important in directing brain plasticity and cognitive function. In other tissue types, disruptions to metabolism and the resultant changes in cellular oxidative state, such as increased reactive oxygen species (ROS) or induction of hypoxia, are associated with cellular stress. In the brain however, where drastic metabolic shifts occur to support physiological processes, subsequent changes to cellular oxidative state and induction of transcriptional sensors of oxidative stress likely play a significant role in regulating physiological neuronal function. Understanding the role of metabolism and metabolically-regulated genes in neuronal function will be critical in elucidating how cognitive functions are disrupted in pathological conditions where neuronal metabolism is affected. Here, we discuss known mechanisms regulating neuronal metabolism as well as the role of hypoxia and oxidative stress during normal and disrupted neuronal function. We also summarize recent studies implicating a role for metabolism in regulating neuronal plasticity as an emerging neuroscience paradigm.
Collapse
Affiliation(s)
- Michelle E Watts
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Charles Claudianos
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia.,Centre for Mental Health Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
21
|
Stykel MG, Humphries K, Kirby MP, Czaniecki C, Wang T, Ryan T, Bamm V, Ryan SD. Nitration of microtubules blocks axonal mitochondrial transport in a human pluripotent stem cell model of Parkinson's disease. FASEB J 2018; 32:5350-5364. [PMID: 29688812 DOI: 10.1096/fj.201700759rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuronal loss in Parkinson's disease (PD) is associated with aberrant mitochondrial function in dopaminergic (DA) neurons of the substantia nigra pars compacta. An association has been reported between PD onset and exposure to mitochondrial toxins, including the agrochemicals paraquat (PQ), maneb (MB), and rotenone (Rot). Here, with the use of a patient-derived stem cell model of PD, allowing comparison of DA neurons harboring a mutation in the α-synuclein (α-syn) gene ( SNCA-A53T) against isogenic, mutation-corrected controls, we describe a novel mechanism whereby NO, generated from SNCA-A53T mutant neurons exposed to Rot or PQ/MB, inhibits anterograde mitochondrial transport through nitration of α-tubulin (α-Tub). Nitration of α-Tub inhibited the association of both α-syn and the mitochondrial motor protein kinesin 5B with the microtubules, arresting anterograde transport. This was, in part, a result of nitration of α-Tub in the C-terminal domain. These effects were rescued by inhibiting NO synthesis with the NOS inhibitor Nω-nitro-L-arginine methyl ester. Collectively, our results are the first to demonstrate a gene by environment interaction in PD, whereby agrochemical exposure selectively triggers a deficit in mitochondrial transport by nitrating the microtubules in neurons harboring the SNCA-A53T mutation.-Stykel, M. G., Humphries, K., Kirby, M. P., Czaniecki, C., Wang, T., Ryan, T., Bamm, V., Ryan, S. D. Nitration of microtubules blocks axonal mitochondrial transport in a human pluripotent stem cell model of Parkinson's disease.
Collapse
Affiliation(s)
- Morgan G Stykel
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Kayla Humphries
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Mathew P Kirby
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Chris Czaniecki
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Tinya Wang
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Tammy Ryan
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Vladimir Bamm
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Scott D Ryan
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.,Neurodegenerative Disease Center, Scintillon Institute, San Diego, California, USA
| |
Collapse
|
22
|
Smita SS, Raj Sammi S, Laxman TS, Bhatta RS, Pandey R. Shatavarin IV elicits lifespan extension and alleviates Parkinsonism in Caenorhabditis elegans. Free Radic Res 2017; 51:954-969. [PMID: 29069955 DOI: 10.1080/10715762.2017.1395419] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Shatavarin IV (SIV), a steroidal saponin, is a major bioactive phytomolecule present in roots of Asparagus racemosus (Liliaceae) known for its anticancer activity. Age-associated neurodegenerative Parkinson's disease (PD) is characterised by alpha-synuclein aggregation in dopaminergic neuron resulting in neurodegeneration. The invention of bioactive molecules that delay aging and age-associated disorders endorses development of natural phytomolecule as a therapeutic agent for curing age-related diseases. Therefore, the present study for the first time explores the potential of SIV against aging and Parkinsonism utilising Caenorhabditis elegans model system. SIV significantly attenuated oxidative stress in terms of intracellular reactive oxygen species (ROS) as well as oxidative damage including protein carbonylation and also promotes longevity. SIV also significantly increased the mRNA expression of stress responsive genes namely sod-1, sod-2, sod-3, gst-4, gst-7 and ctl-2 suggesting its anti-oxidant property that might be contributed in the modulation of oxidative stress and promoting lifespan. Additionally, SIV improved PD symptoms by reducing the alpha-synuclein aggregation, lipid accumulation and enhancing dopamine level. Altogether, present findings indicate that SIV possibly utilising ubiquitin-mediated proteasomal system and attenuating oxidative stress by up-regulating PD-associated genes pdr-1, ubc-12 and pink-1. Therefore, this study is a forward step in exploring the anti-aging and anti-Parkinsonism potential of bioactive compound SIV in C. elegans.
Collapse
Affiliation(s)
- Shachi Shuchi Smita
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| | - Shreesh Raj Sammi
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| | - Tulsankar S Laxman
- b Pharmacokinetics and Metabolism Division , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Rabi S Bhatta
- b Pharmacokinetics and Metabolism Division , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Rakesh Pandey
- a Laboratory of Aging Biology, Department of Microbial Technology and Nematology , Central Institute of Medicinal and Aromatic Plants, Council of Scientific and Industrial Research , Lucknow , India
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The purpose of the study was to discuss the main mechanisms associated with environmental and genetic factors that contribute to the development of Parkinson's disease (PD). RECENT FINDINGS Novel genetic contributors to PD are being identified at a rapid pace in addition to novel environmental factors. The discovery of mutations in alpha-synuclein and leucine-rich repeat kinase 2 causing inherited forms of PD along with epidemiological, in vitro, and in vivo studies identifying herbicides, pesticides, and metals as risk factors have dramatically improved our understanding of mechanisms involved in the development of PD. However, at the same time, these discoveries have also added layers of complexity to the disease. Within the last several years, the genetics associated with PD has dominated the field in many ways; however, the majority of PD cases are likely due to different combinations of environmental exposures and genetic susceptibility. The most common toxicants used to model PD including rotenone, paraquat, and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine have been shown to interact with many of the genes linked with PD such as alpha-synuclein. Therefore, an understanding of mechanisms common between genetic and environmental factors is essential for early detection and successful translation of potential therapies, which is the ultimate goal.
Collapse
Affiliation(s)
- Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, RGE, Rootstown, OH, 44272, USA.
| |
Collapse
|
24
|
Anandhan A, Lei S, Levytskyy R, Pappa A, Panayiotidis MI, Cerny RL, Khalimonchuk O, Powers R, Franco R. Glucose Metabolism and AMPK Signaling Regulate Dopaminergic Cell Death Induced by Gene (α-Synuclein)-Environment (Paraquat) Interactions. Mol Neurobiol 2017. [PMID: 27324791 DOI: 10.1007/s12035-016-9906-2-2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
While environmental exposures are not the single cause of Parkinson's disease (PD), their interaction with genetic alterations is thought to contribute to neuronal dopaminergic degeneration. However, the mechanisms involved in dopaminergic cell death induced by gene-environment interactions remain unclear. In this work, we have revealed for the first time the role of central carbon metabolism and metabolic dysfunction in dopaminergic cell death induced by the paraquat (PQ)-α-synuclein interaction. The toxicity of PQ in dopaminergic N27 cells was significantly reduced by glucose deprivation, inhibition of hexokinase with 2-deoxy-D-glucose (2-DG), or equimolar substitution of glucose with galactose, which evidenced the contribution of glucose metabolism to PQ-induced cell death. PQ also stimulated an increase in glucose uptake, and in the levels of glucose transporter type 4 (GLUT4) and Na+-glucose transporters isoform 1 (SGLT1) proteins, but only inhibition of GLUT-like transport with STF-31 or ascorbic acid reduced PQ-induced cell death. Importantly, while autophagy protein 5 (ATG5)/unc-51 like autophagy activating kinase 1 (ULK1)-dependent autophagy protected against PQ toxicity, the inhibitory effect of glucose deprivation on cell death progression was largely independent of autophagy or mammalian target of rapamycin (mTOR) signaling. PQ selectively induced metabolomic alterations and adenosine monophosphate-activated protein kinase (AMPK) activation in the midbrain and striatum of mice chronically treated with PQ. Inhibition of AMPK signaling led to metabolic dysfunction and an enhanced sensitivity of dopaminergic cells to PQ. In addition, activation of AMPK by PQ was prevented by inhibition of the inducible nitric oxide syntase (iNOS) with 1400W, but PQ had no effect on iNOS levels. Overexpression of wild type or A53T mutant α-synuclein stimulated glucose accumulation and PQ toxicity, and this toxic synergism was reduced by inhibition of glucose metabolism/transport and the pentose phosphate pathway (6-aminonicotinamide). These results demonstrate that glucose metabolism and AMPK regulate dopaminergic cell death induced by gene (α-synuclein)-environment (PQ) interactions.
Collapse
Affiliation(s)
- Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, N200 Beadle Center, Lincoln, NE, 68588-0662, USA
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583-0905, USA
| | - Shulei Lei
- Department of Chemistry, University of Nebraska-Lincoln, Hamilton Hall, Lincoln, NE, 68588-0304, USA
| | - Roman Levytskyy
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0662, USA
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100, Alexandroupolis, Greece
| | | | - Ronald L Cerny
- Department of Chemistry, University of Nebraska-Lincoln, Hamilton Hall, Lincoln, NE, 68588-0304, USA
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0662, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Hamilton Hall, Lincoln, NE, 68588-0304, USA.
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, N200 Beadle Center, Lincoln, NE, 68588-0662, USA.
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583-0905, USA.
| |
Collapse
|
25
|
Paraquat and maneb co-exposure induces noradrenergic locus coeruleus neurodegeneration through NADPH oxidase-mediated microglial activation. Toxicology 2017; 380:1-10. [DOI: 10.1016/j.tox.2017.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/24/2017] [Accepted: 02/09/2017] [Indexed: 01/31/2023]
|
26
|
Truban D, Hou X, Caulfield TR, Fiesel FC, Springer W. PINK1, Parkin, and Mitochondrial Quality Control: What can we Learn about Parkinson's Disease Pathobiology? JOURNAL OF PARKINSON'S DISEASE 2017; 7:13-29. [PMID: 27911343 PMCID: PMC5302033 DOI: 10.3233/jpd-160989] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/10/2016] [Indexed: 12/12/2022]
Abstract
The first clinical description of Parkinson's disease (PD) will embrace its two century anniversary in 2017. For the past 30 years, mitochondrial dysfunction has been hypothesized to play a central role in the pathobiology of this devastating neurodegenerative disease. The identifications of mutations in genes encoding PINK1 (PTEN-induced kinase 1) and Parkin (E3 ubiquitin ligase) in familial PD and their functional association with mitochondrial quality control provided further support to this hypothesis. Recent research focused mainly on their key involvement in the clearance of damaged mitochondria, a process known as mitophagy. It has become evident that there are many other aspects of this complex regulated, multifaceted pathway that provides neuroprotection. As such, numerous additional factors that impact PINK1/Parkin have already been identified including genes involved in other forms of PD. A great pathogenic overlap amongst different forms of familial, environmental and even sporadic disease is emerging that potentially converges at the level of mitochondrial quality control. Tremendous efforts now seek to further detail the roles and exploit PINK1 and Parkin, their upstream regulators and downstream signaling pathways for future translation. This review summarizes the latest findings on PINK1/Parkin-directed mitochondrial quality control, its integration and cross-talk with other disease factors and pathways as well as the implications for idiopathic PD. In addition, we highlight novel avenues for the development of biomarkers and disease-modifying therapies that are based on a detailed understanding of the PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Dominika Truban
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xu Hou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Thomas R. Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Fabienne C. Fiesel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| |
Collapse
|
27
|
Role of sulfiredoxin as a peroxiredoxin-2 denitrosylase in human iPSC-derived dopaminergic neurons. Proc Natl Acad Sci U S A 2016; 113:E7564-E7571. [PMID: 27821734 DOI: 10.1073/pnas.1608784113] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recent studies have pointed to protein S-nitrosylation as a critical regulator of cellular redox homeostasis. For example, S-nitrosylation of peroxiredoxin-2 (Prx2), a peroxidase widely expressed in mammalian neurons, inhibits both enzymatic activity and protective function against oxidative stress. Here, using in vitro and in vivo approaches, we identify a role and reaction mechanism of the reductase sulfiredoxin (Srxn1) as an enzyme that denitrosylates (thus removing -SNO) from Prx2 in an ATP-dependent manner. Accordingly, by decreasing S-nitrosylated Prx2 (SNO-Prx2), overexpression of Srxn1 protects dopaminergic neural cells and human-induced pluripotent stem cell (hiPSC)-derived neurons from NO-induced hypersensitivity to oxidative stress. The pathophysiological relevance of this observation is suggested by our finding that SNO-Prx2 is dramatically increased in murine and human Parkinson's disease (PD) brains. Our findings therefore suggest that Srxn1 may represent a therapeutic target for neurodegenerative disorders such as PD that involve nitrosative/oxidative stress.
Collapse
|
28
|
Nuclear Accumulation of Histone Deacetylase 4 (HDAC4) Exerts Neurotoxicity in Models of Parkinson’s Disease. Mol Neurobiol 2016; 54:6970-6983. [DOI: 10.1007/s12035-016-0199-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/11/2016] [Indexed: 12/18/2022]
|
29
|
Chung SY, Kishinevsky S, Mazzulli JR, Graziotto J, Mrejeru A, Mosharov EV, Puspita L, Valiulahi P, Sulzer D, Milner TA, Taldone T, Krainc D, Studer L, Shim JW. Parkin and PINK1 Patient iPSC-Derived Midbrain Dopamine Neurons Exhibit Mitochondrial Dysfunction and α-Synuclein Accumulation. Stem Cell Reports 2016; 7:664-677. [PMID: 27641647 PMCID: PMC5063469 DOI: 10.1016/j.stemcr.2016.08.012] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 01/29/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the selective loss of dopamine neurons in the substantia nigra; however, the mechanism of neurodegeneration in PD remains unclear. A subset of familial PD is linked to mutations in PARK2 and PINK1, which lead to dysfunctional mitochondria-related proteins Parkin and PINK1, suggesting that pathways implicated in these monogenic forms could play a more general role in PD. We demonstrate that the identification of disease-related phenotypes in PD-patient-specific induced pluripotent stem cell (iPSC)-derived midbrain dopamine (mDA) neurons depends on the type of differentiation protocol utilized. In a floor-plate-based but not a neural-rosette-based directed differentiation strategy, iPSC-derived mDA neurons recapitulate PD phenotypes, including pathogenic protein accumulation, cell-type-specific vulnerability, mitochondrial dysfunction, and abnormal neurotransmitter homeostasis. We propose that these form a pathogenic loop that contributes to disease. Our study illustrates the promise of iPSC technology for examining PD pathogenesis and identifying therapeutic targets.
Collapse
Affiliation(s)
- Sun Young Chung
- Center for Stem Cell Biology, Sloan-Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY 10065, USA
| | - Sarah Kishinevsky
- Center for Stem Cell Biology, Sloan-Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY 10065, USA
| | - Joseph R Mazzulli
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John Graziotto
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ana Mrejeru
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Eugene V Mosharov
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Lesly Puspita
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, 25, Bongjeong-ro, Dongnam-gu, Cheonan-si 31151, Korea
| | - Parvin Valiulahi
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, 25, Bongjeong-ro, Dongnam-gu, Cheonan-si 31151, Korea
| | - David Sulzer
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Tony Taldone
- Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lorenz Studer
- Center for Stem Cell Biology, Sloan-Kettering Institute, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY 10065, USA.
| | - Jae-Won Shim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, 25, Bongjeong-ro, Dongnam-gu, Cheonan-si 31151, Korea.
| |
Collapse
|
30
|
Vidal-Martínez G, Vargas-Medrano J, Gil-Tommee C, Medina D, Garza NT, Yang B, Segura-Ulate I, Dominguez SJ, Perez RG. FTY720/Fingolimod Reduces Synucleinopathy and Improves Gut Motility in A53T Mice: CONTRIBUTIONS OF PRO-BRAIN-DERIVED NEUROTROPHIC FACTOR (PRO-BDNF) AND MATURE BDNF. J Biol Chem 2016; 291:20811-21. [PMID: 27528608 PMCID: PMC5034069 DOI: 10.1074/jbc.m116.744029] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Indexed: 11/06/2022] Open
Abstract
Patients with Parkinson's disease (PD) often have aggregated α-synuclein (aSyn) in enteric nervous system (ENS) neurons, which may be associated with the development of constipation. This occurs well before the onset of classic PD motor symptoms. We previously found that aging A53T transgenic (Tg) mice closely model PD-like ENS aSyn pathology, making them appropriate for testing potential PD therapies. Here we show that Tg mice overexpressing mutant human aSyn develop ENS pathology by 4 months. We then evaluated the responses of Tg mice and their WT littermates to the Food and Drug Administration-approved drug FTY720 (fingolimod, Gilenya) or vehicle control solution from 5 months of age. Long term oral FTY720 in Tg mice reduced ENS aSyn aggregation and constipation, enhanced gut motility, and increased levels of brain-derived neurotrophic factor (BDNF) but produced no significant change in WT littermates. A role for BDNF was directly assessed in a cohort of young A53T mice given vehicle, FTY720, the Trk-B receptor inhibitor ANA-12, or FTY720 + ANA-12 from 1 to 4 months of age. ANA-12-treated Tg mice developed more gut aSyn aggregation as well as constipation, whereas FTY720-treated Tg mice had reduced aSyn aggregation and less constipation, occurring in part by increasing both pro-BDNF and mature BDNF levels. The data from young and old Tg mice revealed FTY720-associated neuroprotection and reduced aSyn pathology, suggesting that FTY720 may also benefit PD patients and others with synucleinopathy. Another finding was a loss of tyrosine hydroxylase immunoreactivity in gut neurons with aggregated aSyn, comparable with our prior findings in the CNS.
Collapse
Affiliation(s)
- Guadalupe Vidal-Martínez
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Javier Vargas-Medrano
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Carolina Gil-Tommee
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - David Medina
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Nathan T Garza
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Barbara Yang
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Ismael Segura-Ulate
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Samantha J Dominguez
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Ruth G Perez
- From the Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas 79905
| |
Collapse
|
31
|
Glucose Metabolism and AMPK Signaling Regulate Dopaminergic Cell Death Induced by Gene (α-Synuclein)-Environment (Paraquat) Interactions. Mol Neurobiol 2016; 54:3825-3842. [PMID: 27324791 DOI: 10.1007/s12035-016-9906-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
While environmental exposures are not the single cause of Parkinson's disease (PD), their interaction with genetic alterations is thought to contribute to neuronal dopaminergic degeneration. However, the mechanisms involved in dopaminergic cell death induced by gene-environment interactions remain unclear. In this work, we have revealed for the first time the role of central carbon metabolism and metabolic dysfunction in dopaminergic cell death induced by the paraquat (PQ)-α-synuclein interaction. The toxicity of PQ in dopaminergic N27 cells was significantly reduced by glucose deprivation, inhibition of hexokinase with 2-deoxy-D-glucose (2-DG), or equimolar substitution of glucose with galactose, which evidenced the contribution of glucose metabolism to PQ-induced cell death. PQ also stimulated an increase in glucose uptake, and in the levels of glucose transporter type 4 (GLUT4) and Na+-glucose transporters isoform 1 (SGLT1) proteins, but only inhibition of GLUT-like transport with STF-31 or ascorbic acid reduced PQ-induced cell death. Importantly, while autophagy protein 5 (ATG5)/unc-51 like autophagy activating kinase 1 (ULK1)-dependent autophagy protected against PQ toxicity, the inhibitory effect of glucose deprivation on cell death progression was largely independent of autophagy or mammalian target of rapamycin (mTOR) signaling. PQ selectively induced metabolomic alterations and adenosine monophosphate-activated protein kinase (AMPK) activation in the midbrain and striatum of mice chronically treated with PQ. Inhibition of AMPK signaling led to metabolic dysfunction and an enhanced sensitivity of dopaminergic cells to PQ. In addition, activation of AMPK by PQ was prevented by inhibition of the inducible nitric oxide syntase (iNOS) with 1400W, but PQ had no effect on iNOS levels. Overexpression of wild type or A53T mutant α-synuclein stimulated glucose accumulation and PQ toxicity, and this toxic synergism was reduced by inhibition of glucose metabolism/transport and the pentose phosphate pathway (6-aminonicotinamide). These results demonstrate that glucose metabolism and AMPK regulate dopaminergic cell death induced by gene (α-synuclein)-environment (PQ) interactions.
Collapse
|
32
|
Choi J, Polcher A, Joas A. Systematic literature review on Parkinson's disease and Childhood Leukaemia and mode of actions for pesticides. ACTA ACUST UNITED AC 2016. [DOI: 10.2903/sp.efsa.2016.en-955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Wan N, Lin G. Parkinson's Disease and Pesticides Exposure: New Findings From a Comprehensive Study in Nebraska, USA. J Rural Health 2015; 32:303-13. [PMID: 26515233 DOI: 10.1111/jrh.12154] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND The association between exposure to agricultural pesticides and Parkinson's Disease (PD) has long been a topic of study in the field of environmental health. This research takes advantage of the unique Nebraska PD registry and state-level crop classification data to investigate the PD-pesticides exposure relationship. METHODS First, Geographic Information System and satellite remote sensing data were adopted to calculate exposure to different pesticides for Nebraska residents. An integrated spatial exploratory framework was then adopted to explore the association between PD incidence and exposure to specific pesticide ingredients at the county level. RESULTS Our results reveal similarities in geographic patterns of pesticide exposure and PD incidence. The regression analyses indicate that, for most Nebraska counties, PD incidence was significantly associated with exposure to certain pesticide ingredients such as alachlor and broxomy. However, the results also suggest that factors other than pesticide exposure may help further explain the risk of PD at the county level. CONCLUSIONS We found significant associations between PD incidence and exposure to different pesticide ingredients. These results have useful implications for PD prevention in Nebraska and other agricultural states in the United States.
Collapse
Affiliation(s)
- Neng Wan
- Department of Geography, University of Utah, Salt Lake City, Utah
| | - Ge Lin
- School of Community Health Sciences, University of Nevada - Las Vegas, Las Vegas, Nevada
| |
Collapse
|
34
|
Electron Transport Disturbances and Neurodegeneration: From Albert Szent-Györgyi's Concept (Szeged) till Novel Approaches to Boost Mitochondrial Bioenergetics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:498401. [PMID: 26301042 PMCID: PMC4537740 DOI: 10.1155/2015/498401] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
Impaired function of certain mitochondrial respiratory complexes has long been linked to the pathogenesis of chronic neurodegenerative disorders such as Parkinson's and Huntington's diseases. Furthermore, genetic alterations of mitochondrial genome or nuclear genes encoding proteins playing essential roles in maintaining proper mitochondrial function can lead to the development of severe systemic diseases associated with neurodegeneration and vacuolar myelinopathy. At present, all of these diseases lack effective disease modifying therapy. Following a brief commemoration of Professor Albert Szent-Györgyi, a Nobel Prize laureate who pioneered in the field of cellular respiration, antioxidant processes, and the roles of free radicals in health and disease, the present paper overviews the current knowledge on the involvement of mitochondrial dysfunction in central nervous system diseases associated with neurodegeneration including Parkinson's and Huntington's disease as well as mitochondrial encephalopathies. The review puts special focus on the involvement and the potential therapeutic relevance of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α), a nuclear-encoded master regulator of mitochondrial biogenesis and antioxidant responses in these disorders, the transcriptional activation of which may hold novel therapeutic value as a more system-based approach aiming to restore mitochondrial functions in neurodegenerative processes.
Collapse
|
35
|
Okamoto SI, Ambasudhan R, Nakamura T, Lipton SA. S-Nitrosylation of MEF2: a common mechanism underlying neurological disorders? FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.14.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Shu-ichi Okamoto
- Neuroscience & Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Rajesh Ambasudhan
- Neuroscience & Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Tomohiro Nakamura
- Neuroscience & Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neuroscience & Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Neurosciences, University of California SanDiego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92048, USA
| |
Collapse
|
36
|
Pan-Montojo F, Reichmann H. Considerations on the role of environmental toxins in idiopathic Parkinson's disease pathophysiology. Transl Neurodegener 2014; 3:10. [PMID: 24826210 PMCID: PMC4019355 DOI: 10.1186/2047-9158-3-10] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/29/2014] [Indexed: 12/21/2022] Open
Abstract
Neurodegenerative diseases are characterized by a progressive dysfunction of the nervous system. Often associated with atrophy of the affected central or peripheral nervous structures, they include diseases such as Parkinson’s Disease (PD), Alzheimer’s Disease and other dementias, Genetic Brain Disorders, Amyotrophic Lateral Sclerosis (ALS or Lou Gehrig’s Disease), Huntington’s Disease, Prion Diseases, and others. The prevalence of neurodegenerative diseases has increased over the last years. This has had a major impact both on patients and their families and has exponentially increased the medical bill by hundreds of billions of Euros. Therefore, understanding the role of environmental and genetic factors in the pathogenesis of PD is crucial to develop preventive strategies. While some authors believe that PD is mainly genetic and that the aging of the society is the principal cause for this increase, different studies suggest that PD may be due to an increased exposure to environmental toxins. In this article we review epidemiological, sociological and experimental studies to determine which hypothesis is more plausible. Our conclusion is that, at least in idiopathic PD (iPD), the exposure to toxic environmental substances could play an important role in its aetiology.
Collapse
Affiliation(s)
- Francisco Pan-Montojo
- Neurologische Klinik, Klinikum der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany ; Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut Ludwig-Maximilians-Universität München, Schillerstr. 44, 80336 Munich, Germany
| | - Heinz Reichmann
- Klinik und Poliklinik für Neurologie, Carl Gustav Carus University Hospital, TU-Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
37
|
Camilleri A, Vassallo N. The centrality of mitochondria in the pathogenesis and treatment of Parkinson's disease. CNS Neurosci Ther 2014; 20:591-602. [PMID: 24703487 DOI: 10.1111/cns.12264] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/07/2014] [Accepted: 03/08/2014] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disorder leading to progressive motor impairment and for which there is no cure. From the first postmortem account describing a lack of mitochondrial complex I in the substantia nigra of PD sufferers, the direct association between mitochondrial dysfunction and death of dopaminergic neurons has ever since been consistently corroborated. In this review, we outline common pathways shared by both sporadic and familial PD that remarkably and consistently converge at the level of mitochondrial integrity. Furthermore, such knowledge has incontrovertibly established mitochondria as a valid therapeutic target in neurodegeneration. We discuss several mitochondria-directed therapies that promote the preservation, rescue, or restoration of dopaminergic neurons and which have been identified in the laboratory and in preclinical studies. Some of these have progressed to clinical trials, albeit the identification of an unequivocal disease-modifying neurotherapeutic is still elusive. The challenge is therefore to improve further, not least by more research on the molecular mechanisms and pathophysiological consequences of mitochondrial dysfunction in PD.
Collapse
Affiliation(s)
- Angelique Camilleri
- Department of Physiology and Biochemistry, University of Malta, Msida 2080, Malta
| | | |
Collapse
|
38
|
Nuber S, Tadros D, Fields J, Overk CR, Ettle B, Kosberg K, Mante M, Rockenstein E, Trejo M, Masliah E. Environmental neurotoxic challenge of conditional alpha-synuclein transgenic mice predicts a dopaminergic olfactory-striatal interplay in early PD. Acta Neuropathol 2014; 127:477-94. [PMID: 24509835 DOI: 10.1007/s00401-014-1255-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/31/2014] [Accepted: 02/01/2014] [Indexed: 12/17/2022]
Abstract
The olfactory bulb (OB) is one of the first brain regions in Parkinson's disease (PD) to contain alpha-synuclein (α-syn) inclusions, possibly associated with nonmotor symptoms. Mechanisms underlying olfactory synucleinopathy, its contribution to progressive aggregation pathology and nigrostriatal dopaminergic loss observed at later stages, remain unclear. A second hit, such as environmental toxins, is suggestive for α-syn aggregation in olfactory neurons, potentially triggering disease progression. To address the possible pathogenic role of olfactory α-syn accumulation in early PD, we exposed mice with site-specific and inducible overexpression of familial PD-linked mutant α-syn in OB neurons to a low dose of the herbicide paraquat. Here, we found that olfactory α-syn per se elicited structural and behavioral abnormalities, characteristic of an early time point in models with widespread α-syn expression, including hyperactivity and increased striatal dopaminergic marker. Suppression of α-syn reversed the dopaminergic phenotype. In contrast, paraquat treatment synergistically induced degeneration of olfactory dopaminergic cells and opposed the higher reactive phenotype. Neither neurodegeneration nor behavioral abnormalities were detected in paraquat-treated mice with suppressed α-syn expression. By increasing calpain activity, paraquat induced a pathological cascade leading to inhibition of autophagy clearance and accumulation of calpain-cleaved truncated and insoluble α-syn, recapitulating biochemical and structural changes in human PD. Thus our results underscore the primary role of proteolytic failure in aggregation pathology. In addition, we provide novel evidence that olfactory dopaminergic neurons display an increased vulnerability toward neurotoxins in dependence to presence of human α-syn, possibly mediating an olfactory-striatal dopaminergic network dysfunction in mouse models and early PD.
Collapse
Affiliation(s)
- Silke Nuber
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., MTF 344, La Jolla, CA, 92093-0624, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Isogenic human iPSC Parkinson's model shows nitrosative stress-induced dysfunction in MEF2-PGC1α transcription. Cell 2013; 155:1351-64. [PMID: 24290359 DOI: 10.1016/j.cell.2013.11.009] [Citation(s) in RCA: 345] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/11/2013] [Accepted: 11/04/2013] [Indexed: 01/16/2023]
Abstract
Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.
Collapse
|
40
|
Perfeito R, Cunha-Oliveira T, Rego AC. Reprint of: revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease-resemblance to the effect of amphetamine drugs of abuse. Free Radic Biol Med 2013; 62:186-201. [PMID: 23743292 DOI: 10.1016/j.freeradbiomed.2013.05.042] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a chronic and progressive neurological disease associated with a loss of dopaminergic neurons. In most cases the disease is sporadic but genetically inherited cases also exist. One of the major pathological features of PD is the presence of aggregates that localize in neuronal cytoplasm as Lewy bodies, mainly composed of α-synuclein (α-syn) and ubiquitin. The selective degeneration of dopaminergic neurons suggests that dopamine itself may contribute to the neurodegenerative process in PD. Furthermore, mitochondrial dysfunction and oxidative stress constitute key pathogenic events of this disorder. Thus, in this review we give an actual perspective to classical pathways involving these two mechanisms of neurodegeneration, including the role of dopamine in sporadic and familial PD, as well as in the case of abuse of amphetamine-type drugs. Mutations in genes related to familial PD causing autosomal dominant or recessive forms may also have crucial effects on mitochondrial morphology, function, and oxidative stress. Environmental factors, such as MPTP and rotenone, have been reported to induce selective degeneration of the nigrostriatal pathways leading to α-syn-positive inclusions, possibly by inhibiting mitochondrial complex I of the respiratory chain and subsequently increasing oxidative stress. Recently, increased risk for PD was found in amphetamine users. Amphetamine drugs have effects similar to those of other environmental factors for PD, because long-term exposure to these drugs leads to dopamine depletion. Moreover, amphetamine neurotoxicity involves α-syn aggregation, mitochondrial dysfunction, and oxidative stress. Therefore, dopamine and related oxidative stress, as well as mitochondrial dysfunction, seem to be common links between PD and amphetamine neurotoxicity.
Collapse
Affiliation(s)
- Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| |
Collapse
|
41
|
Sunico CR, Nakamura T, Rockenstein E, Mante M, Adame A, Chan SF, Newmeyer TF, Masliah E, Nakanishi N, Lipton SA. S-Nitrosylation of parkin as a novel regulator of p53-mediated neuronal cell death in sporadic Parkinson's disease. Mol Neurodegener 2013; 8:29. [PMID: 23985028 PMCID: PMC3765907 DOI: 10.1186/1750-1326-8-29] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022] Open
Abstract
Background Mutations in the gene encoding parkin, a neuroprotective protein with dual functions as an E3 ubiquitin ligase and transcriptional repressor of p53, are linked to familial forms of Parkinson’s disease (PD). We hypothesized that oxidative posttranslational modification of parkin by environmental toxins may contribute to sporadic PD. Results We first demonstrated that S-nitrosylation of parkin decreased its activity as a repressor of p53 gene expression, leading to upregulation of p53. Chromatin immunoprecipitation as well as gel-shift assays showed that parkin bound to the p53 promoter, and this binding was inhibited by S-nitrosylation of parkin. Additionally, nitrosative stress induced apoptosis in cells expressing parkin, and this death was, at least in part, dependent upon p53. In primary mesencephalic cultures, pesticide-induced apoptosis was prevented by inhibition of nitric oxide synthase (NOS). In a mouse model of pesticide-induced PD, both S-nitrosylated (SNO-)parkin and p53 protein levels were increased, while administration of a NOS inhibitor mitigated neuronal death in these mice. Moreover, the levels of SNO-parkin and p53 were simultaneously elevated in postmortem human PD brain compared to controls. Conclusions Taken together, our data indicate that S-nitrosylation of parkin, leading to p53-mediated neuronal cell death, contributes to the pathophysiology of sporadic PD.
Collapse
Affiliation(s)
- Carmen R Sunico
- Sanford-Burnham Medical Research Institute, Del E, Webb Center for Neuroscience, Aging, and Stem Cell Research, 10901, North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bichler Z, Lim HC, Zeng L, Tan EK. Non-motor and motor features in LRRK2 transgenic mice. PLoS One 2013; 8:e70249. [PMID: 23936174 PMCID: PMC3728021 DOI: 10.1371/journal.pone.0070249] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 06/18/2013] [Indexed: 12/13/2022] Open
Abstract
Background Non-motor symptoms are increasingly recognized as important features of Parkinson’s disease (PD). LRRK2 mutations are common causes of familial and sporadic PD. Non-motor features have not been yet comprehensively evaluated in LRRK2 transgenic mouse models. Objective Using a transgenic mouse model overexpressing the R1441G mutation of the human LRRK2 gene, we have investigated the longitudinal correlation between motor and non-motor symptoms and determined if specific non-motor phenotypes precede motor symptoms. Methodology We investigated the onset of motor and non-motor phenotypes on the LRRK2R1441G BAC transgenic mice and their littermate controls from 4 to 21 month-old using a battery of behavioral tests. The transgenic mutant mice displayed mild hypokinesia in the open field from 16 months old, with gastrointestinal dysfunctions beginning at 6 months old. Non-motor features such as depression and anxiety-like behaviors, sensorial functions (pain sensitivity and olfaction), and learning and memory abilities in the passive avoidance test were similar in the transgenic animals compared to littermate controls. Conclusions LRRK2R1441G BAC transgenic mice displayed gastrointestinal dysfunction at an early stage but did not have abnormalities in fine behaviors, olfaction, pain sensitivity, mood disorders and learning and memory compared to non-transgenic littermate controls. The observations on olfaction and gastrointestinal dysfunction in this model validate findings in human carriers. These mice did recapitulate mild Parkinsonian motor features at late stages but compensatory mechanisms modulating the progression of PD in these models should be further evaluated.
Collapse
Affiliation(s)
- Zoë Bichler
- Behavioral Neuroscience Laboratory, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Graduate Medical School, Program in Neuroscience and Behavioral Disorders, Singapore, Singapore
- * E-mail: (ZB); (EKT)
| | - Han Chi Lim
- Neural Stem Cell Laboratory, National Neuroscience Institute, Singapore, Singapore
| | - Li Zeng
- Duke-NUS Graduate Medical School, Program in Neuroscience and Behavioral Disorders, Singapore, Singapore
- Neural Stem Cell Laboratory, National Neuroscience Institute, Singapore, Singapore
| | - Eng King Tan
- Behavioral Neuroscience Laboratory, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Graduate Medical School, Program in Neuroscience and Behavioral Disorders, Singapore, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
- * E-mail: (ZB); (EKT)
| |
Collapse
|
43
|
Subramaniam SR, Chesselet MF. Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog Neurobiol 2013; 106-107:17-32. [PMID: 23643800 PMCID: PMC3742021 DOI: 10.1016/j.pneurobio.2013.04.004] [Citation(s) in RCA: 563] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/13/2013] [Accepted: 04/22/2013] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a movement disorder that is characterized by the progressive degeneration of dopaminergic neurons in substantia nigra pars compacta resulting in dopamine deficiency in the striatum. Although majority of the PD cases are sporadic several genetic mutations have also been linked to the disease thus providing new opportunities to study the pathology of the illness. Studies in humans and various animal models of PD reveal that mitochondrial dysfunction might be a defect that occurs early in PD pathogenesis and appears to be a widespread feature in both sporadic and monogenic forms of PD. The general mitochondrial abnormalities linked with the disease include mitochondrial electron transport chain impairment, alterations in mitochondrial morphology and dynamics, mitochondrial DNA mutations and anomaly in calcium homeostasis. Mitochondria are vital organelles with multiple functions and their dysfunction can lead to a decline in energy production, generation of reactive oxygen species and induction of stress-induced apoptosis. In this review, we give an outline of mitochondrial functions that are affected in the pathogenesis of sporadic and familial PD, and hence provide insights that might be valuable for focused future research to exploit possible mitochondrial targets for neuroprotective interventions in PD.
Collapse
Affiliation(s)
- Sudhakar Raja Subramaniam
- Department of Neurology, David Geffen School of Medicine, UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | | |
Collapse
|
44
|
Chorfa A, Bétemps D, Morignat E, Lazizzera C, Hogeveen K, Andrieu T, Baron T. Specific pesticide-dependent increases in α-synuclein levels in human neuroblastoma (SH-SY5Y) and melanoma (SK-MEL-2) cell lines. Toxicol Sci 2013; 133:289-97. [PMID: 23535362 DOI: 10.1093/toxsci/kft076] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Epidemiological studies indicate a role of genetic and environmental factors in Parkinson's disease involving alterations of the neuronal α-synuclein (α-syn) protein. In particular, a relationship between Parkinson's disease and occupational exposure to pesticides has been repeatedly suggested. Our objective was to precisely assess changes in α-syn levels in human neuroblastoma (SH-SY5Y) and melanoma (SK-MEL-2) cell lines following acute exposure to pesticides (rotenone, paraquat, maneb, and glyphosate) using Western blot and flow cytometry. These human cell lines express α-syn endogenously, and overexpression of α-syn (wild type or mutated A53T) can be obtained following recombinant adenoviral transduction. We found that endogenous α-syn levels in the SH-SY5Y neuroblastoma cell line were markedly increased by paraquat, and to a lesser extent by rotenone and maneb, but not by glyphosate. Rotenone also clearly increased endogenous α-syn levels in the SK-MEL-2 melanoma cell line. In the SH-SY5Y cell line, similar differences were observed in the α-syn adenovirus-transduced cells, with a higher increase of the A53T mutated protein. Paraquat markedly increased α-syn in the SK-MEL-2 adenovirus-transduced cell line, similarly for the wild-type or A53T proteins. The observed differences in the propensities of pesticides to increase α-syn levels are in agreement with numerous reports that indicate a potential role of exposure to certain pesticides in the development of Parkinson's disease. Our data support the hypothesis that pesticides can trigger some molecular events involved in this disease and also in malignant melanoma that consistently shows a significant but still unexplained association with Parkinson's disease.
Collapse
Affiliation(s)
- Areski Chorfa
- Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (Anses), Unité Maladies Neuro-Dégénératives, 69394 Lyon Cedex 07, France
| | | | | | | | | | | | | |
Collapse
|
45
|
Deleersnijder A, Gerard M, Debyser Z, Baekelandt V. The remarkable conformational plasticity of alpha-synuclein: blessing or curse? Trends Mol Med 2013; 19:368-77. [DOI: 10.1016/j.molmed.2013.04.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 04/03/2013] [Accepted: 04/03/2013] [Indexed: 12/21/2022]
|
46
|
Oaks AW, Frankfurt M, Finkelstein DI, Sidhu A. Age-dependent effects of A53T alpha-synuclein on behavior and dopaminergic function. PLoS One 2013; 8:e60378. [PMID: 23560093 PMCID: PMC3613356 DOI: 10.1371/journal.pone.0060378] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 02/25/2013] [Indexed: 11/30/2022] Open
Abstract
Expression of A53T mutant human alpha-synuclein under the mouse prion promoter is among the most successful transgenic models of Parkinson's disease. Accumulation of A53T alpha-synuclein causes adult mice to develop severe motor impairment resulting in early death at 8–12 months of age. In younger, pre-symptomatic animals, altered motor activity and anxiety-like behaviors have also been reported. These behavioral changes, which precede severe neuropathology, may stem from non-pathological functions of alpha-synuclein, including modulation of monoamine neurotransmission. Our analysis over the adult life-span of motor activity, anxiety-like, and depressive-like behaviors identifies perturbations both before and after the onset of disease. Young A53T mice had increased distribution of the dopamine transporter (DAT) to the membrane that was associated with increased striatal re-uptake function. DAT function decreased with aging, and was associated with neurochemical alterations that included increased expression of beta-synuclein and gamma synuclein. Prior to normalization of dopamine uptake, transient activation of Tau kinases and hyperphosphorylation of Tau in the striatum were also observed. Aged A53T mice had reduced neuron counts in the substantia nigra pars compacta, yet striatal medium spiny neuron dendritic spine density was largely maintained. These findings highlight the involvement of the synuclein family of proteins and phosphorylation of Tau in the response to dopaminergic dysfunction of the nigrostriatal pathway.
Collapse
Affiliation(s)
- Adam W. Oaks
- Laboratory of Molecular Neurochemistry, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Maya Frankfurt
- Department of Science Education, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, United States of America
| | - David I. Finkelstein
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anita Sidhu
- Laboratory of Molecular Neurochemistry, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
47
|
Chung SJ, Armasu SM, Anderson KJ, Biernacka JM, Lesnick TG, Rider DN, Cunningham JM, Ahlskog JE, Frigerio R, Maraganore DM. Genetic susceptibility loci, environmental exposures, and Parkinson's disease: a case-control study of gene-environment interactions. Parkinsonism Relat Disord 2013; 19:595-9. [PMID: 23507417 DOI: 10.1016/j.parkreldis.2013.02.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Revised: 01/16/2013] [Accepted: 02/15/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND Prior studies causally linked mutations in SNCA, MAPT, and LRRK2 genes with familial Parkinsonism. Genome-wide association studies have demonstrated association of single nucleotide polymorphisms (SNPs) in those three genes with sporadic Parkinson's disease (PD) susceptibility worldwide. Here we investigated the interactions between SNPs in those three susceptibility genes and environmental exposures (pesticides application, tobacco smoking, coffee drinking, and alcohol drinking) also associated with PD susceptibility. METHODS Pairwise interactions between environmental exposures and 18 variants (16 SNPs and two variable number tandem repeats, or "VNTRs") in SNCA, MAPT and LRRK2, were investigated using data from 1098 PD cases from the upper Midwest, USA and 1098 matched controls. Environmental exposures were assessed using a validated telephone interview script. RESULTS Five pairwise interactions had uncorrected P-values < 0.05. These included pairings of pesticides × SNCA rs3775423 or MAPT rs4792891, coffee drinking × MAPT H1/H2 haplotype or MAPT rs16940806, and alcohol drinking × MAPT rs2435211. None of these interactions remained significant after Bonferroni correction. Secondary analyses in strata defined by type of control (sibling or unrelated), sex, or age at onset of the case also did not identify significant interactions after Bonferroni correction. CONCLUSIONS This study documented limited pairwise interactions between established genetic and environmental risk factors for PD; however, the associations were not significant after correction for multiple testing.
Collapse
Affiliation(s)
- Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Perfeito R, Cunha-Oliveira T, Rego AC. Revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease--resemblance to the effect of amphetamine drugs of abuse. Free Radic Biol Med 2012; 53:1791-806. [PMID: 22967820 DOI: 10.1016/j.freeradbiomed.2012.08.569] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a chronic and progressive neurological disease associated with a loss of dopaminergic neurons. In most cases the disease is sporadic but genetically inherited cases also exist. One of the major pathological features of PD is the presence of aggregates that localize in neuronal cytoplasm as Lewy bodies, mainly composed of α-synuclein (α-syn) and ubiquitin. The selective degeneration of dopaminergic neurons suggests that dopamine itself may contribute to the neurodegenerative process in PD. Furthermore, mitochondrial dysfunction and oxidative stress constitute key pathogenic events of this disorder. Thus, in this review we give an actual perspective to classical pathways involving these two mechanisms of neurodegeneration, including the role of dopamine in sporadic and familial PD, as well as in the case of abuse of amphetamine-type drugs. Mutations in genes related to familial PD causing autosomal dominant or recessive forms may also have crucial effects on mitochondrial morphology, function, and oxidative stress. Environmental factors, such as MPTP and rotenone, have been reported to induce selective degeneration of the nigrostriatal pathways leading to α-syn-positive inclusions, possibly by inhibiting mitochondrial complex I of the respiratory chain and subsequently increasing oxidative stress. Recently, increased risk for PD was found in amphetamine users. Amphetamine drugs have effects similar to those of other environmental factors for PD, because long-term exposure to these drugs leads to dopamine depletion. Moreover, amphetamine neurotoxicity involves α-syn aggregation, mitochondrial dysfunction, and oxidative stress. Therefore, dopamine and related oxidative stress, as well as mitochondrial dysfunction, seem to be common links between PD and amphetamine neurotoxicity.
Collapse
Affiliation(s)
- Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | | |
Collapse
|
49
|
Silva BA, Breydo L, Fink AL, Uversky VN. Agrochemicals, α-synuclein, and Parkinson's disease. Mol Neurobiol 2012; 47:598-612. [PMID: 22933040 DOI: 10.1007/s12035-012-8333-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 08/13/2012] [Indexed: 12/21/2022]
Abstract
Epidemiological, population-based case-control, and experimental studies at the molecular, cellular, and organism levels revealed that exposure to various environmental agents, including a number of structurally different agrochemicals, may contribute to the pathogenesis of Parkinson's disease (PD) and several other neurodegenerative disorders. The role of genetic predisposition in PD has also been increasingly acknowledged, driven by the identification of a number of disease-related genes [e.g., α-synuclein, parkin, DJ-1, ubiquitin C-terminal hydrolase isozyme L1 (UCH-L1), and nuclear receptor-related factor 1]. Therefore, the etiology of this multifactorial disease is likely to involve both genetic and environmental factors. Various neurotoxicants, including agrochemicals, have been shown to elevate the levels of α-synuclein expression in neurons and to promote aggregation of this protein in vivo. Many agrochemicals physically interact with α-synuclein and accelerate the fibrillation and aggregation rates of this protein in vitro. This review analyzes some of the aspects linking α-synuclein to PD, provides brief structural and functional descriptions of this important protein, and represents some data connecting exposure to agrochemicals with α-synuclein aggregation and PD pathogenesis.
Collapse
Affiliation(s)
- Blanca A Silva
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA
| | | | | | | |
Collapse
|
50
|
Butler EK, Voigt A, Lutz AK, Toegel JP, Gerhardt E, Karsten P, Falkenburger B, Reinartz A, Winklhofer KF, Schulz JB. The mitochondrial chaperone protein TRAP1 mitigates α-Synuclein toxicity. PLoS Genet 2012; 8:e1002488. [PMID: 22319455 PMCID: PMC3271059 DOI: 10.1371/journal.pgen.1002488] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 12/02/2011] [Indexed: 01/13/2023] Open
Abstract
Overexpression or mutation of α-Synuclein is associated with protein aggregation and interferes with a number of cellular processes, including mitochondrial integrity and function. We used a whole-genome screen in the fruit fly Drosophila melanogaster to search for novel genetic modifiers of human [A53T]α-Synuclein-induced neurotoxicity. Decreased expression of the mitochondrial chaperone protein tumor necrosis factor receptor associated protein-1 (TRAP1) was found to enhance age-dependent loss of fly head dopamine (DA) and DA neuron number resulting from [A53T]α-Synuclein expression. In addition, decreased TRAP1 expression in [A53T]α-Synuclein-expressing flies resulted in enhanced loss of climbing ability and sensitivity to oxidative stress. Overexpression of human TRAP1 was able to rescue these phenotypes. Similarly, human TRAP1 overexpression in rat primary cortical neurons rescued [A53T]α-Synuclein-induced sensitivity to rotenone treatment. In human (non)neuronal cell lines, small interfering RNA directed against TRAP1 enhanced [A53T]α-Synuclein-induced sensitivity to oxidative stress treatment. [A53T]α-Synuclein directly interfered with mitochondrial function, as its expression reduced Complex I activity in HEK293 cells. These effects were blocked by TRAP1 overexpression. Moreover, TRAP1 was able to prevent alteration in mitochondrial morphology caused by [A53T]α-Synuclein overexpression in human SH-SY5Y cells. These results indicate that [A53T]α-Synuclein toxicity is intimately connected to mitochondrial dysfunction and that toxicity reduction in fly and rat primary neurons and human cell lines can be achieved using overexpression of the mitochondrial chaperone TRAP1. Interestingly, TRAP1 has previously been shown to be phosphorylated by the serine/threonine kinase PINK1, thus providing a potential link of PINK1 via TRAP1 to α-Synuclein.
Collapse
Affiliation(s)
- Erin K. Butler
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biology (GGNB), Göttingen, Germany
| | - Aaron Voigt
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - A. Kathrin Lutz
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
| | - Jane P. Toegel
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Ellen Gerhardt
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
| | - Peter Karsten
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Björn Falkenburger
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Andrea Reinartz
- Department of Pathology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Konstanze F. Winklhofer
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jörg B. Schulz
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Jülich-Aachen Research Alliance (JARA) Brain, Jülich/Aachen, Germany
- * E-mail:
| |
Collapse
|