1
|
Lee SH, Tonello R, Lee K, Roh J, Prudente AS, Kim YH, Park CK, Berta T. The Parkinson's disease DJ-1/PARK7 gene controls peripheral neuronal excitability and painful neuropathy. Brain 2025; 148:1639-1651. [PMID: 39486088 PMCID: PMC12073980 DOI: 10.1093/brain/awae341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disease with well-documented motor symptoms and less recognized, but significant, non-motor symptoms. These non-motor symptoms include prodromal pain and peripheral neuropathy, the causes of which are unknown. We investigated the role of DJ-1/PARK7, a Parkinson's disease-associated gene, in prodromal pain and peripheral neuropathy. Using Dj-1-deficient mice, we conducted comprehensive sensory tests, cutaneous staining, molecular analyses and electrophysiological studies on mouse and human primary sensory neurons from dorsal root ganglia. We found that these mice exhibited cold hypersensitivity, oxidative stress, and neuropathy of the cutaneous fibres of primary sensory neurons before any motor impairments were observed. Mechanistically, DJ-1 in primary sensory neurons regulated this hypersensitivity and neuropathy via TRPA1 signalling. Interestingly, we discovered that DJ-1 also plays a role in the progression of chemotherapy-induced peripheral neuropathies. Pain and mechanisms associated with these neuropathies were exacerbated in Dj-1-deficient mice but were significantly reduced by the pharmacological activation of Dj-1. Importantly, we also confirmed the expression of DJ-1 and its therapeutic potential in human primary sensory neurons. Thus, we uncover a peripheral mechanism of DJ-1 and propose that it might serve as a new target for developing therapeutic approaches for Parkinson's disease-linked and other painful neuropathies.
Collapse
Affiliation(s)
- Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York, NY 10010, USA
| | - Kihwan Lee
- Tooth-Periodontium Complex Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Jueun Roh
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| |
Collapse
|
2
|
Mishra R, Upadhyay A. An update on mammalian and non-mammalian animal models for biomarker development in neurodegenerative disorders. Cell Mol Life Sci 2025; 82:147. [PMID: 40192808 PMCID: PMC11977071 DOI: 10.1007/s00018-025-05668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Neurodegeneration is one of the leading factor for death globally, affecting millions of people. Developing animal models are critical to understand biological processes and comprehend pathological hallmarks of neurodegenerative diseases. For decades, many animal models have served as excellent tools to determine the disease progression, develop diagnostic methods and design novel therapies against distinct pathologies. Here, we provide a comprehensive overview of both, mammalian and non-mammalian animal models, with a focus on three most common and aggressive neurodegenerative disorders: Alzheimer's disease, Parkinson's disease and Spinocerebellar ataxia-1. We highlight various approaches including transgene, gene transfer, and chemically-induced methods used to develop disease models. In particular, we discuss applications of both non-mammalian and mammalian contributions in research on neurodegeneration. It is exciting to learn the roles of animal models in disease pathomechanisms, identifying biomarkers and hence devising novel interventions to treat neuropathological conditions.
Collapse
Affiliation(s)
- Ribhav Mishra
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, 491002, India
| |
Collapse
|
3
|
Lim D, Matute C, Cavaliere F, Verkhratsky A. Neuroglia in neurodegeneration: Alzheimer, Parkinson, and Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:9-44. [PMID: 40148060 DOI: 10.1016/b978-0-443-19102-2.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The conspicuous rise of chronic neurodegenerative diseases, including Alzheimer (AD), Parkinson (PD), and Huntington (HD) diseases, is currently without disease-modifying therapies and accompanied by an excessive rate of unsuccessful clinical trials. This reflects a profound lack of understanding of the pathogenesis of these diseases, indicating that the current paradigms guiding disease modeling and drug development are in need of reconsideration. The role of neuroglia, namely astrocytes, microglial cells, and oligodendrocytes, in the pathogenesis of neurodegenerative diseases emerged during the last decades. This chapter provides the state-of-the-art update on the changes of astrocytes, microglial cells, and oligodendrocytes in AD, PD, and HD. A growing body of evidence suggests that homeostatic and defensive functions of glial cells are compromised at different disease stages, leading to increased susceptibility of neurons to noxious stimuli, eventually resulting in their malfunction and degeneration. Investments are needed in the generation of novel preclinical models suitable for studying glial pathology, in "humanizing" research, and in-depth investigation of glial cell alterations to slow down and, possibly, halt and prevent the rise of neurodegenerative disease. Targeting glial cells opens new therapeutic avenues to treat AD, PD, and HD.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy.
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain
| | - Fabio Cavaliere
- The Basque Biomodels Platform for Human Research (BBioH), Achucarro Basque Center for Neuroscience & Fundación Biofisica Bizkaia, Leioa, Spain
| | - Alexei Verkhratsky
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
4
|
Rostad KO, Trognitz T, Frøyset AK, Bifulco E, Fladmark KE. Accelerated Sarcopenia Phenotype in the DJ-1/ Park7-Knockout Zebrafish. Antioxidants (Basel) 2024; 13:1509. [PMID: 39765837 PMCID: PMC11673048 DOI: 10.3390/antiox13121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Age-dependent loss of muscle mass and function is associated with oxidative stress. DJ-1/Park7 acts as an antioxidant through multiple signalling pathways. DJ-1-knockout zebrafish show a decline in swimming performance and loss of weight gain between 6 and 9 months of age. Here, we address the degree to which this is associated with muscle degeneration and identify molecular changes preceding dysregulation of muscle performance. Loss of DJ-1 reduced the skeletal muscle fibre cross-section area. The highly mitochondrial-dependent red slow muscle was more affected than the white muscle, and degeneration of sub-sarcolemma red muscle mitochondria was observed. Using TandemMassTag-based quantitative proteomics, we identified a total of 3721 proteins in the multiplex sample of 4 and 12-month-old muscles. A total of 68 proteins, mainly associated with inflammation and mitochondrial function, were dysregulated in the young DJ-1-null adults, with Annexin A3, Sphingomyelin phosphodiesterase acid-like 3B, Complement C3a, and 2,4-dienoyl CoA reductase 1 being the most affected. The loss of DJ-1 also accelerated molecular features associated with sarcopenia, such as a decrease in the NAD+/NADH ratio and a reduction in Prostaglandin reductase 2 and Cytosolic glycerol-3-phosphate dehydrogenase levels. In view of the experimental power of zebrafish, the DJ-1-null zebrafish makes a valuable model for understanding the connection between oxidative stress and age-dependent muscle loss and function.
Collapse
Affiliation(s)
| | | | | | | | - Kari E. Fladmark
- Department of Biological Sciences, University of Bergen, 5020 Bergen, Norway (T.T.); (A.K.F.); (E.B.)
| |
Collapse
|
5
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [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: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
6
|
Guimarães RP, de Resende MCS, Tavares MM, Belardinelli de Azevedo C, Ruiz MCM, Mortari MR. Construct, Face, and Predictive Validity of Parkinson's Disease Rodent Models. Int J Mol Sci 2024; 25:8971. [PMID: 39201659 PMCID: PMC11354451 DOI: 10.3390/ijms25168971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally. Current drugs only alleviate symptoms without halting disease progression, making rodent models essential for researching new therapies and understanding the disease better. However, selecting the right model is challenging due to the numerous models and protocols available. Key factors in model selection include construct, face, and predictive validity. Construct validity ensures the model replicates pathological changes seen in human PD, focusing on dopaminergic neurodegeneration and a-synuclein aggregation. Face validity ensures the model's symptoms mirror those in humans, primarily reproducing motor and non-motor symptoms. Predictive validity assesses if treatment responses in animals will reflect those in humans, typically involving classical pharmacotherapies and surgical procedures. This review highlights the primary characteristics of PD and how these characteristics are validated experimentally according to the three criteria. Additionally, it serves as a valuable tool for researchers in selecting the most appropriate animal model based on established validation criteria.
Collapse
Affiliation(s)
- Rayanne Poletti Guimarães
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Maria Clara Souza de Resende
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Mesquita Tavares
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Caio Belardinelli de Azevedo
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Cesar Merino Ruiz
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
- Neurological Rehabilitation Unit, Sarah Network of Rehabilitation Hospitals, Brasília 70335-901, Brazil
| | - Márcia Renata Mortari
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| |
Collapse
|
7
|
Lind-Holm Mogensen F, Sousa C, Ameli C, Badanjak K, Pereira SL, Muller A, Kaoma T, Coowar D, Scafidi A, Poovathingal SK, Tziortziou M, Antony PMA, Nicot N, Ginolhac A, Vogt Weisenhorn DM, Wurst W, Poli A, Nazarov PV, Skupin A, Grünewald A, Michelucci A. PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation. J Neuroinflammation 2024; 21:174. [PMID: 39014482 PMCID: PMC11253405 DOI: 10.1186/s12974-024-03164-x] [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: 02/05/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation. METHODS Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs). RESULTS By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs. CONCLUSIONS Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Carole Sousa
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Corrado Ameli
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Katja Badanjak
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Sandro L Pereira
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Djalil Coowar
- Rodent Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Suresh K Poovathingal
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Single Cell Analytics and Microfluidics Core, Vlaams Instituut Voor Biotechnologie-KU Leuven, 3000, Louvain, Belgium
| | - Maria Tziortziou
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Paul M A Antony
- Bioimaging Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Daniela M Vogt Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Deutsche Zentrum für Psychische Gesundheit (DZPG), 80336, Munich, Germany
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Petr V Nazarov
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Multiomics Data Science Group, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Alexander Skupin
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Neuroscience, University of California San Diego, La Jolla, CA, 92093, USA
- Integrative Biophysics, Department of Physics and Material Science, University of Luxembourg, L-1511, Luxembourg, Luxembourg
| | - Anne Grünewald
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
8
|
Harraz MM. Selective dopaminergic vulnerability in Parkinson's disease: new insights into the role of DAT. Front Neurosci 2023; 17:1219441. [PMID: 37694119 PMCID: PMC10483232 DOI: 10.3389/fnins.2023.1219441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023] Open
Abstract
One of the hallmarks of Parkinson's disease (PD) is the progressive loss of dopaminergic neurons and associated dopamine depletion. Several mechanisms, previously considered in isolation, have been proposed to contribute to the pathophysiology of dopaminergic degeneration: dopamine oxidation-mediated neurotoxicity, high dopamine transporter (DAT) expression density per neuron, and autophagy-lysosome pathway (ALP) dysfunction. However, the interrelationships among these mechanisms remained unclear. Our recent research bridges this gap, recognizing autophagy as a novel dopamine homeostasis regulator, unifying these concepts. I propose that autophagy modulates dopamine reuptake by selectively degrading DAT. In PD, ALP dysfunction could increase DAT density per neuron, and enhance dopamine reuptake, oxidation, and neurotoxicity, potentially contributing to the progressive loss of dopaminergic neurons. This integrated understanding may provide a more comprehensive view of aspects of PD pathophysiology and opens new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Maged M. Harraz
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
10
|
Dovonou A, Bolduc C, Soto Linan V, Gora C, Peralta Iii MR, Lévesque M. Animal models of Parkinson's disease: bridging the gap between disease hallmarks and research questions. Transl Neurodegener 2023; 12:36. [PMID: 37468944 PMCID: PMC10354932 DOI: 10.1186/s40035-023-00368-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still provide valuable information that contributes to our understanding of the disease and the limitations of our treatment options. This review comprehensively summarizes the different animal models available for Parkinson's research, with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to realize that the strengths and weaknesses of each model and the induction technique at our disposal are determined by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete discernment of classical and novel animal models in PD research.
Collapse
Affiliation(s)
- Axelle Dovonou
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Cyril Bolduc
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Victoria Soto Linan
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Charles Gora
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Modesto R Peralta Iii
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
11
|
Khan E, Hasan I, Haque ME. Parkinson's Disease: Exploring Different Animal Model Systems. Int J Mol Sci 2023; 24:ijms24109088. [PMID: 37240432 DOI: 10.3390/ijms24109088] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Disease modeling in non-human subjects is an essential part of any clinical research. To gain proper understanding of the etiology and pathophysiology of any disease, experimental models are required to replicate the disease process. Due to the huge diversity in pathophysiology and prognosis in different diseases, animal modeling is customized and specific accordingly. As in other neurodegenerative diseases, Parkinson's disease is a progressive disorder coupled with varying forms of physical and mental disabilities. The pathological hallmarks of Parkinson's disease are associated with the accumulation of misfolded protein called α-synuclein as Lewy body, and degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc) area affecting the patient's motor activity. Extensive research has already been conducted regarding animal modeling of Parkinson's diseases. These include animal systems with induction of Parkinson's, either pharmacologically or via genetic manipulation. In this review, we will be summarizing and discussing some of the commonly employed Parkinson's disease animal model systems and their applications and limitations.
Collapse
Affiliation(s)
- Engila Khan
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ikramul Hasan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - M Emdadul Haque
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
12
|
Lind-Holm Mogensen F, Scafidi A, Poli A, Michelucci A. PARK7/DJ-1 in microglia: implications in Parkinson's disease and relevance as a therapeutic target. J Neuroinflammation 2023; 20:95. [PMID: 37072827 PMCID: PMC10111685 DOI: 10.1186/s12974-023-02776-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/11/2023] [Indexed: 04/20/2023] Open
Abstract
Microglia are the immune effector cells of the brain playing critical roles in immune surveillance and neuroprotection in healthy conditions, while they can sustain neuroinflammatory and neurotoxic processes in neurodegenerative diseases, including Parkinson's disease (PD). Although the precise triggers of PD remain obscure, causative genetic mutations, which aid in the identification of molecular pathways underlying the pathogenesis of idiopathic forms, represent 10% of the patients. Among the inherited forms, loss of function of PARK7, which encodes the protein DJ-1, results in autosomal recessive early-onset PD. Yet, although protection against oxidative stress is the most prominent task ascribed to DJ-1, the underlying mechanisms linking DJ-1 deficiency to the onset of PD are a current matter of investigation. This review provides an overview of the role of DJ-1 in neuroinflammation, with a special focus on its functions in microglia genetic programs and immunological traits. Furthermore, it discusses the relevance of targeting dysregulated pathways in microglia under DJ-1 deficiency and their importance as therapeutic targets in PD. Lastly, it addresses the prospect to consider DJ-1, detected in its oxidized form in idiopathic PD, as a biomarker and to take into account DJ-1-enhancing compounds as therapeutics dampening oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Doctoral School of Science and Technology, University of Luxembourg, 7 Avenue Des Haut Forneuaux, L-4362, Esch-Sur-Alzette, Luxembourg
| | - Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Doctoral School of Science and Technology, University of Luxembourg, 7 Avenue Des Haut Forneuaux, L-4362, Esch-Sur-Alzette, Luxembourg
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
13
|
Chavali LNM, Yddal I, Bifulco E, Mannsåker S, Røise D, Law JO, Frøyset AK, Grellscheid SN, Fladmark KE. Progressive Motor and Non-Motor Symptoms in Park7 Knockout Zebrafish. Int J Mol Sci 2023; 24:ijms24076456. [PMID: 37047429 PMCID: PMC10094626 DOI: 10.3390/ijms24076456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
DJ-1 is a redox sensitive protein with a wide range of functions related to oxidative stress protection. Mutations in the park7 gene, which codes for DJ-1 are associated with early onset familial Parkinson’s disease and increased astrocytic DJ-1 levels are found in pathologic tissues from idiopathic Parkinson’s disease. We have previously established a DJ-1 knockout zebrafish line that developed normally, but with aging the DJ-1 null fish had a lowered level of tyrosine hydroxylase, respiratory mitochondrial failure and a lower body mass. Here we have examined the DJ-1 knockout from the early adult stage and show that loss of DJ-1 results in a progressive, age-dependent increase in both motoric and non-motoric symptoms associated to Parkinson’s disease. These changes coincide with changes in mitochondrial and mitochondrial associated proteins. Recent studies have suggested that a decline in NAD+ can contribute to Parkinson’s disease and that supplementation of NAD+ precursors may delay disease progression. We found that the brain NAD+/NADH ratio decreased in aging zebrafish but did not correlate with DJ-1 induced altered behavior. Differences were first observed at the late adult stage in which NAD+ and NADPH levels were decreased in DJ-1 knockouts. Considering the experimental power of zebrafish and the development of Parkinson’s disease-related symptoms in the DJ-1 null fish, this model can serve as a useful tool both to understand the progression of the disease and the effect of suggested treatments.
Collapse
|
14
|
Jimenez-Harrison D, Huseby CJ, Hoffman CN, Sher S, Snyder D, Seal B, Yuan C, Fu H, Wysocki V, Giorgini F, Kuret J. DJ-1 Molecular Chaperone Activity Depresses Tau Aggregation Propensity through Interaction with Monomers. Biochemistry 2023; 62:976-988. [PMID: 36813261 PMCID: PMC9997487 DOI: 10.1021/acs.biochem.2c00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/19/2023] [Indexed: 02/24/2023]
Abstract
Tau aggregate-bearing lesions are pathological markers and potential mediators of tauopathic neurodegenerative diseases, including Alzheimer's disease. The molecular chaperone DJ-1 colocalizes with tau pathology in these disorders, but it has been unclear what functional link exists between them. In this study, we examined the consequences of tau/DJ-1 interaction as isolated proteins in vitro. When added to full-length 2N4R tau under aggregation-promoting conditions, DJ-1 inhibited both the rate and extent of filament formation in a concentration-dependent manner. Inhibitory activity was low affinity, did not require ATP, and was not affected by substituting oxidation incompetent missense mutation C106A for wild-type DJ-1. In contrast, missense mutations previously linked to familial Parkinson's disease and loss of α-synuclein chaperone activity, M26I and E64D, displayed diminished tau chaperone activity relative to wild-type DJ-1. Although DJ-1 directly bound the isolated microtubule-binding repeat region of tau protein, exposure of preformed tau seeds to DJ-1 did not diminish seeding activity in a biosensor cell model. These data reveal DJ-1 to be a holdase chaperone capable of engaging tau as a client in addition to α-synuclein. Our findings support a role for DJ-1 as part of an endogenous defense against the aggregation of these intrinsically disordered proteins.
Collapse
Affiliation(s)
- Daniela Jimenez-Harrison
- Medical
Scientist Training Program, The Ohio State
University, Columbus, Ohio 43210, United States
| | - Carol J. Huseby
- Department
of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, Ohio 43210, United States
| | - Claire N. Hoffman
- Department
of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, Ohio 43210, United States
| | - Steven Sher
- Medical
Scientist Training Program, The Ohio State
University, Columbus, Ohio 43210, United States
| | - Dalton Snyder
- Department
of Chemistry and Biochemistry, The Ohio
State University College of Medicine, Columbus, Ohio 43210, United States
| | - Brayden Seal
- Department
of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, Ohio 43210, United States
| | - Chunhua Yuan
- Campus
Chemical Instrument Center, The Ohio State
University College of Medicine, Columbus, Ohio 43210, United States
| | - Hongjun Fu
- Department
of Neuroscience, The Ohio State University
College of Medicine, Columbus, Ohio 43210, United States
| | - Vicki Wysocki
- Department
of Chemistry and Biochemistry, The Ohio
State University College of Medicine, Columbus, Ohio 43210, United States
| | - Flaviano Giorgini
- Department
of Genetics and Genome Biology, University
of Leicester, Leicester LE1 7RH, United
Kingdom
| | - Jeff Kuret
- Department
of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, Ohio 43210, United States
| |
Collapse
|
15
|
A novel splicing variant of DJ-1 in Parkinson's disease induces mitochondrial dysfunction. Heliyon 2023; 9:e14039. [PMID: 36915530 PMCID: PMC10006478 DOI: 10.1016/j.heliyon.2023.e14039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
Several studies have identified mutations in neuroprotective genes in a few cases of Parkinson's disease (PD); however, the role of alternative splicing changes in PD remains unelucidated. Based on the transcriptome analysis of substantia nigra (SN) tissues obtained from PD cases and age-matched healthy controls, we identified a novel alternative splicing variant of DJ-1, lacking exon 6 (DJ-1 ΔE6), frequently detected in the SN of patients with PD. We found that the exon 6 skipping of DJ-1 induces mitochondrial dysfunction and impaired antioxidant capability. According to an in silico modeling study, the exon 6 skipping of DJ-1 disrupts the structural state suitable for the oxidation of the cysteine 106 residue that is a prerequisite for activating its neuroprotective roles. Our results suggest that change in DJ-1 alternative splicing may contribute to PD progression and provide an insight for studying PD etiology and its potential therapeutic targets.
Collapse
|
16
|
Zhang C, Chen S, Li X, Xu Q, Lin Y, Lin F, Yuan M, Zi Y, Cai J. Progress in Parkinson's disease animal models of genetic defects: Characteristics and application. Biomed Pharmacother 2022; 155:113768. [DOI: 10.1016/j.biopha.2022.113768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/15/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
|
17
|
Kim J, Daadi EW, Oh T, Daadi ES, Daadi MM. Human Induced Pluripotent Stem Cell Phenotyping and Preclinical Modeling of Familial Parkinson's Disease. Genes (Basel) 2022; 13:1937. [PMID: 36360174 PMCID: PMC9689743 DOI: 10.3390/genes13111937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022] Open
Abstract
Parkinson's disease (PD) is primarily idiopathic and a highly heterogenous neurodegenerative disease with patients experiencing a wide array of motor and non-motor symptoms. A major challenge for understanding susceptibility to PD is to determine the genetic and environmental factors that influence the mechanisms underlying the variations in disease-associated traits. The pathological hallmark of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta region of the brain and post-mortem Lewy pathology, which leads to the loss of projecting axons innervating the striatum and to impaired motor and cognitive functions. While the cause of PD is still largely unknown, genome-wide association studies provide evidence that numerous polymorphic variants in various genes contribute to sporadic PD, and 10 to 15% of all cases are linked to some form of hereditary mutations, either autosomal dominant or recessive. Among the most common mutations observed in PD patients are in the genes LRRK2, SNCA, GBA1, PINK1, PRKN, and PARK7/DJ-1. In this review, we cover these PD-related mutations, the use of induced pluripotent stem cells as a disease in a dish model, and genetic animal models to better understand the diversity in the pathogenesis and long-term outcomes seen in PD patients.
Collapse
Affiliation(s)
- Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
| | - Etienne W. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Elyas S. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Marcel M. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
- Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
18
|
Chen W, Liu H, Liu S, Kang Y, Nie Z, Lei H. Altered prefrontal neurochemistry in the DJ-1 knockout mouse model of Parkinson's disease: complementary semi-quantitative analyses with in vivo magnetic resonance spectroscopy and MALDI-MSI. Anal Bioanal Chem 2022; 414:7977-7987. [PMID: 36208327 DOI: 10.1007/s00216-022-04341-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022]
Abstract
In vivo proton magnetic resonance spectroscopy (1H-MRS) and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) are two semi-quantitative analytical methods commonly used in neurochemical research. In this study, the two methods were used complementarily, in parallel, to investigate neurochemical perturbations in the medial prefrontal cortex (mPFC) of 9-month-old DJ-1 knockout mice, a well-established transgenic model for Parkinson's diseases. Convergingly, the results obtained with the two methods demonstrated that, compared with the wild-type (WT) mice, the DJ-1 knockout mice had significantly increased glutathione (GSH) level and GSH/glutamate (Glu) ratio in the mPFC, which likely presented an astrocytic compensatory mechanism in response to elevated regional oxidative stress induced by the loss of DJ-1 function. The results from this study also highlighted (1) the need to be cautious when interpreting the in vivo 1H-MRS results obtained from aged transgenic animals, in which the concentration of internal reference, being whether water or total creatine, could no longer be assumed to be the same as that in the age-matched WT animals, and (2) the necessity and importance of complementary analyses with more than one method under such circumstances.
Collapse
Affiliation(s)
- Wei Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Huihui Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China
| | - Sijie Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yan Kang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China
| | - Zongxiu Nie
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China. .,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China.
| | - Hao Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China. .,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
19
|
Ghosh A, Mustafiz A, Pareek A, Sopory SK, Singla-Pareek SL. Glyoxalase III enhances salinity tolerance through reactive oxygen species scavenging and reduced glycation. PHYSIOLOGIA PLANTARUM 2022; 174:e13693. [PMID: 35483971 DOI: 10.1111/ppl.13693] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 06/14/2023]
Abstract
Methylglyoxal (MG) is a metabolically generated highly cytotoxic compound that accumulates in all living organisms, from Escherichia coli to humans, under stress conditions. To detoxify MG, nature has evolved reduced glutathione (GSH)-dependent glyoxalase and NADPH-dependent aldo-keto reductase systems. But both GSH and NADPH have been reported to be limiting in plants under stress conditions, and thus detoxification might not be performed efficiently. Recently, glyoxalase III (GLY III)-like enzyme activity has been reported from various species, which can detoxify MG without any cofactor. In the present study, we have tested whether an E. coli gene, hchA, encoding a functional GLY III, could provide abiotic stress tolerance to living systems. Overexpression of this gene showed improved tolerance in E. coli and Saccharomyces cerevisiae cells against salinity, dicarbonyl, and oxidative stresses. Ectopic expression of the E. coli GLY III gene (EcGLY-III) in transgenic tobacco plants confers tolerance against salinity at both seedling and reproductive stages as indicated by their height, weight, membrane stability index, and total yield potential. Transgenic plants showed significantly increased glyoxalase and antioxidant enzyme activity that resisted the accumulation of excess MG and reactive oxygen species (ROS) during stress. Moreover, transgenic plants showed more anti-glycation activity to inhibit the formation of advanced glycation end product (AGE) that might prevent transgenic plants from stress-induced senescence. Taken together, all these observations indicate that overexpression of EcGLYIII confers salinity stress tolerance in plants and should be explored further for the generation of stress-tolerant plants.
Collapse
Affiliation(s)
- Ajit Ghosh
- Plant Stress Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ananda Mustafiz
- Plant Stress Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ashwani Pareek
- Stress Physiology and Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sudhir K Sopory
- Plant Stress Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sneh L Singla-Pareek
- Plant Stress Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
20
|
Klonarakis M, De Vos M, Woo E, Ralph L, Thacker JS, Gil-Mohapel J. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neurosci Biobehav Rev 2022; 135:104541. [DOI: 10.1016/j.neubiorev.2022.104541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/28/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
|
21
|
Lama J, Buhidma Y, Fletcher E, Duty S. Animal models of Parkinson's disease: a guide to selecting the optimal model for your research. Neuronal Signal 2021; 5:NS20210026. [PMID: 34956652 PMCID: PMC8661507 DOI: 10.1042/ns20210026] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a complex, multisystem disorder characterised by α-synuclein (SNCA) pathology, degeneration of nigrostriatal dopaminergic neurons, multifactorial pathogenetic mechanisms and expression of a plethora of motor and non-motor symptoms. Animal models of PD have already been instructive in helping us unravel some of these aspects. However, much remains to be discovered, requiring continued interrogation by the research community. In contrast with the situation for many neurological disorders, PD benefits from of a wide range of available animal models (pharmacological, toxin, genetic and α-synuclein) but this makes selection of the optimal one for a given study difficult. This is especially so when a study demands a model that displays a specific combination of features. While many excellent reviews of animal models already exist, this review takes a different approach with the intention of more readily informing this decision-making process. We have considered each feature of PD in turn - aetiology, pathology, pathogenesis, motor dysfunctions and non-motor symptoms (NMS) - highlighting those animal models that replicate each. By compiling easily accessible tables and a summary figure, we aim to provide the reader with a simple, go-to resource for selecting the optimal animal model of PD to suit their research needs.
Collapse
Affiliation(s)
- Joana Lama
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Yazead Buhidma
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Edward J.R. Fletcher
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Susan Duty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| |
Collapse
|
22
|
Bastioli G, Regoni M, Cazzaniga F, De Luca CMG, Bistaffa E, Zanetti L, Moda F, Valtorta F, Sassone J. Animal Models of Autosomal Recessive Parkinsonism. Biomedicines 2021; 9:biomedicines9070812. [PMID: 34356877 PMCID: PMC8301401 DOI: 10.3390/biomedicines9070812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder. The neuropathological hallmark of the disease is the loss of dopamine neurons of the substantia nigra pars compacta. The clinical manifestations of PD are bradykinesia, rigidity, resting tremors and postural instability. PD patients often display non-motor symptoms such as depression, anxiety, weakness, sleep disturbances and cognitive disorders. Although, in 90% of cases, PD has a sporadic onset of unknown etiology, highly penetrant rare genetic mutations in many genes have been linked with typical familial PD. Understanding the mechanisms behind the DA neuron death in these Mendelian forms may help to illuminate the pathogenesis of DA neuron degeneration in the more common forms of PD. A key step in the identification of the molecular pathways underlying DA neuron death, and in the development of therapeutic strategies, is the creation and characterization of animal models that faithfully recapitulate the human disease. In this review, we outline the current status of PD modeling using mouse, rat and non-mammalian models, focusing on animal models for autosomal recessive PD.
Collapse
Affiliation(s)
- Guendalina Bastioli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Maria Regoni
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Federico Cazzaniga
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Chiara Maria Giulia De Luca
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Edoardo Bistaffa
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Letizia Zanetti
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Fabio Moda
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (F.C.); (C.M.G.D.L.); (E.B.); (F.M.)
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Jenny Sassone
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (G.B.); (M.R.); (L.Z.); (F.V.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence:
| |
Collapse
|
23
|
Huang M, Chen S. DJ-1 in neurodegenerative diseases: Pathogenesis and clinical application. Prog Neurobiol 2021; 204:102114. [PMID: 34174373 DOI: 10.1016/j.pneurobio.2021.102114] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/22/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (NDs) are one of the major health threats to human characterized by selective and progressive neuronal loss. The mechanisms of NDs are still not fully understood. The study of genetic defects and disease-related proteins offers us a window into the mystery of it, and the extension of knowledge indicates that different NDs share similar features, mechanisms, and even genetic or protein abnormalities. Among these findings, PARK7 and its production DJ-1 protein, which was initially found implicated in PD, have also been found altered in other NDs. PARK7 mutations, altered expression and posttranslational modification (PTM) cause DJ-1 abnormalities, which in turn lead to downstream mechanisms shared by most NDs, such as mitochondrial dysfunction, oxidative stress, protein aggregation, autophagy defects, and so on. The knowledge of DJ-1 derived from PD researches might apply to other NDs in both basic research and clinical application, and might yield novel insights into and alternative approaches for dealing with NDs.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China; Lab for Translational Research of Neurodegenerative Diseases, Institute of Immunochemistry, Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
24
|
Airavaara M, Parkkinen I, Konovalova J, Albert K, Chmielarz P, Domanskyi A. Back and to the Future: From Neurotoxin-Induced to Human Parkinson's Disease Models. ACTA ACUST UNITED AC 2020; 91:e88. [PMID: 32049438 DOI: 10.1002/cpns.88] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by motor symptoms such as tremor, slowness of movement, rigidity, and postural instability, as well as non-motor features like sleep disturbances, loss of ability to smell, depression, constipation, and pain. Motor symptoms are caused by depletion of dopamine in the striatum due to the progressive loss of dopamine neurons in the substantia nigra pars compacta. Approximately 10% of PD cases are familial arising from genetic mutations in α-synuclein, LRRK2, DJ-1, PINK1, parkin, and several other proteins. The majority of PD cases are, however, idiopathic, i.e., having no clear etiology. PD is characterized by progressive accumulation of insoluble inclusions, known as Lewy bodies, mostly composed of α-synuclein and membrane components. The cause of PD is currently attributed to cellular proteostasis deregulation and mitochondrial dysfunction, which are likely interdependent. In addition, neuroinflammation is present in brains of PD patients, but whether it is the cause or consequence of neurodegeneration remains to be studied. Rodents do not develop PD or PD-like motor symptoms spontaneously; however, neurotoxins, genetic mutations, viral vector-mediated transgene expression and, recently, injections of misfolded α-synuclein have been successfully utilized to model certain aspects of the disease. Here, we critically review the advantages and drawbacks of rodent PD models and discuss approaches to advance pre-clinical PD research towards successful disease-modifying therapy. © 2020 The Authors.
Collapse
Affiliation(s)
- Mikko Airavaara
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Julia Konovalova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Katrina Albert
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Trinh D, Israwi AR, Arathoon LR, Gleave JA, Nash JE. The multi-faceted role of mitochondria in the pathology of Parkinson's disease. J Neurochem 2020; 156:715-752. [PMID: 33616931 DOI: 10.1111/jnc.15154] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria are essential for neuronal function. They produce ATP to meet energy demands, regulate homeostasis of ion levels such as calcium and regulate reactive oxygen species that cause oxidative cellular stress. Mitochondria have also been shown to regulate protein synthesis within themselves, as well as within the nucleus, and also influence synaptic plasticity. These roles are especially important for neurons, which have higher energy demands and greater susceptibility to stress. Dysfunction of mitochondria has been associated with several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, Huntington's disease, Glaucoma and Amyotrophic Lateral Sclerosis. The focus of this review is on how and why mitochondrial function is linked to the pathology of Parkinson's disease (PD). Many of the PD-linked genetic mutations which have been identified result in dysfunctional mitochondria, through a wide-spread number of mechanisms. In this review, we describe how susceptible neurons are predisposed to be vulnerable to the toxic events that occur during the neurodegenerative process of PD, and how mitochondria are central to these pathways. We also discuss ways in which proteins linked with familial PD control mitochondrial function, both physiologically and pathologically, along with their implications in genome-wide association studies and risk assessment. Finally, we review potential strategies for disease modification through mitochondrial enhancement. Ultimately, agents capable of both improving and/or restoring mitochondrial function, either alone, or in conjunction with other disease-modifying agents may halt or slow the progression of neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Dennison Trinh
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Ahmad R Israwi
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Lindsay R Arathoon
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Jacqueline A Gleave
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Joanne E Nash
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| |
Collapse
|
26
|
Li M, Xu H, Chen G, Sun S, Wang Q, Liu B, Wu X, Zhou L, Chai Z, Sun X, Lu Y, Younus M, Zheng L, Zhu F, Jia H, Chen X, Wang C, Zhou Z. Impaired D2 receptor-dependent dopaminergic transmission in prefrontal cortex of awake mouse model of Parkinson's disease. Brain 2020; 142:3099-3115. [PMID: 31504219 DOI: 10.1093/brain/awz243] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/02/2019] [Accepted: 06/19/2019] [Indexed: 12/27/2022] Open
Abstract
The loss-of-function mutation in PARK7/DJ-1 is one of the most common causes of autosomal recessive Parkinson's disease, and patients carrying PARK7 mutations often exhibit both a progressive movement disorder and emotional impairment, such as anxiety. However, the causes of the emotional symptom accompanying PARK7-associated and other forms of Parkinson's disease remain largely unexplored. Using two-photon microscopic Ca2+ imaging in awake PARK7-/- and PARK7+/+ mice, we found that (i) PARK7-/- neurons in the frontal association cortex showed substantially higher circuit activity recorded as spontaneous somatic Ca2+ signals; (ii) both basal and evoked dopamine release remained intact, as determined by both electrochemical dopamine recordings and high performance liquid chromatography in vivo; (iii) D2 receptor expression was significantly decreased in postsynaptic frontal association cortical neurons, and the hyper-neuronal activity were rescued by D2 receptor intervention using either local pharmacology or viral D2 receptor over-expression; and (iv) PARK7-/- mice showed anxiety-like behaviours that were rescued by either local D2 receptor pharmacology or overexpression. Thus, for first time, we demonstrated a robust D2 receptor-dependent phenotype of individual neurons within the prefrontal cortex circuit in awake parkinsonian mice that linked with anxiety. Our work sheds light on early-onset phenotypes and the mechanisms underlying Parkinson's disease by imaging brain circuits in an awake mouse model.
Collapse
Affiliation(s)
- Mingli Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Huadong Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.,Key Lab of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Guoqing Chen
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Xi Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yang Lu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Muhammad Younus
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Lianghong Zheng
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Hongbo Jia
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xiaowei Chen
- Brain Research Center, Third Military Medical University, Chongqing, China
| | - Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.,Center for Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
27
|
White D, de Sousa Abreu RP, Blake A, Murphy J, Showell S, Kitamoto T, Lawal HO. Deficits in the vesicular acetylcholine transporter alter lifespan and behavior in adult Drosophila melanogaster. Neurochem Int 2020; 137:104744. [PMID: 32315665 DOI: 10.1016/j.neuint.2020.104744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
Abstract
The neurotransmitter acetylcholine (ACh) is involved in critical organismal functions that include locomotion and cognition. Importantly, alterations in the cholinergic system are a key underlying factor in cognitive defects associated with aging. One essential component of cholinergic synaptic transmission is the vesicular ACh transporter (VAChT), which regulates the packaging of ACh into synaptic vesicles for extracellular release. Mutations that cause a reduction in either protein level or activity lead to diminished locomotion ability whereas complete loss of function of VAChT is lethal. While much is known about the function of VAChT, the direct role of altered ACh release and its association with either an impairment or an enhancement of cognitive function are still not fully understood. We hypothesize that point mutations in Vacht cause age-related deficits in cholinergic-mediated behaviors such as locomotion, and learning and memory. Using Drosophila melanogaster as a model system, we have studied several mutations within Vacht and observed their effect on survivability and locomotive behavior. Here we report for the first time a weak hypomorphic Vacht allele that shows a differential effect on ACh-linked behaviors. We also demonstrate that partially rescued Vacht point mutations cause an allele-dependent deficit in lifespan and defects in locomotion ability. Moreover, using a thorough data analytics strategy to identify exploratory behavioral patterns, we introduce new paradigms for measuring locomotion-related activities that could not be revealed or detected by a simple measure of the average speed alone. Together, our data indicate a role for VAChT in the maintenance of longevity and locomotion abilities in Drosophila and we provide additional measurements of locomotion that can be useful in determining subtle changes in Vacht function on locomotion-related behaviors.
Collapse
Affiliation(s)
- Daniel White
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Raquel P de Sousa Abreu
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Andrew Blake
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Jeremy Murphy
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Shardae Showell
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Toshihiro Kitamoto
- Department of Anesthesia, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Hakeem O Lawal
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA.
| |
Collapse
|
28
|
Historical Perspective: Models of Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21072464. [PMID: 32252301 PMCID: PMC7177377 DOI: 10.3390/ijms21072464] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is the most common movement disorder with motor and nonmotor signs. The current therapeutic regimen for PD is mainly symptomatic as the etio-pathophysiology has not been fully elucidated. A variety of animal models has been generated to study different aspects of the disease for understanding the pathogenesis and therapeutic development. The disease model can be generated through neurotoxin-based or genetic-based approaches in a wide range of animals such as non-human primates (NHP), rodents, zebrafish, Caenorhabditis (C.) elegans, and drosophila. Cellular-based disease model is frequently used because of the ease of manipulation and suitability for large-screen assays. In neurotoxin-induced models, chemicals such as 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, and paraquat are used to recapitulate the disease. Genetic manipulation of PD-related genes, such as α-Synuclein(SNCA), Leucine-rich repeat kinase 2 (LRRK2), Pten-Induced Kinase 1 (PINK1), Parkin(PRKN), and Protein deglycase (DJ-1) Are used in the transgenic models. An emerging model that combines both genetic- and neurotoxin-based methods has been generated to study the role of the immune system in the pathogenesis of PD. Here, we discuss the advantages and limitations of the different PD models and their utility for different research purposes.
Collapse
|
29
|
Choi DJ, Yang H, Gaire S, Lee KA, An J, Kim BG, Jou I, Park SM, Joe EH. Critical roles of astrocytic-CCL2-dependent monocyte infiltration in a DJ-1 knockout mouse model of delayed brain repair. Glia 2020; 68:2086-2101. [PMID: 32176388 DOI: 10.1002/glia.23828] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/25/2020] [Accepted: 02/27/2020] [Indexed: 01/01/2023]
Abstract
Monocyte-derived macrophages play a role in the repair of the injured brain. We previously reported that a deficiency of the Parkinson's disease (PD)-associated gene DJ-1 delays repair of brain injury produced by stereotaxic injection of ATP, a component of damage-associated molecular patterns. Here, we show that a DJ-1 deficiency attenuates monocyte infiltration into the damaged brain owing to a decrease in C-C motif chemokine ligand 2 (CCL2) expression in astrocytes. Like DJ-1-knockout (KO) mice, CCL2 receptor (CCR2)-KO mice showed defects in monocyte infiltration and delayed recovery of brain injury, as determined by 9.4 T magnetic resonance imaging analysis and immunostaining for tyrosine hydroxylase and glial fibrillary acid protein. Notably, transcriptome analyses showed that genes related to regeneration and synapse formation were similarly downregulated in injured brains of DJ-1-KO and CCR2-KO mice compared with the injured wild-type brain. These results indicate that defective astrogliosis in DJ-1-KO mice is associated with decreased CCL2 expression and attenuated monocyte infiltration, resulting in delayed repair of brain injury. Thus, delayed repair of brain injury could contribute to the development of PD. MAIN POINTS: A DJ-1 deficiency attenuates infiltration of monocytes owing to a decrease in CCL2 expression in astrocytes, which in turn led to delay in repair of brain injury.
Collapse
Affiliation(s)
- Dong-Joo Choi
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Hajie Yang
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Sushil Gaire
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Keon Ah Lee
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Jiawei An
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Byung Gon Kim
- Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Neurology, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Ilo Jou
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Sang Myun Park
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| | - Eun-Hye Joe
- Department of Pharmacology, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Kyunggi-do, South Korea
| |
Collapse
|
30
|
Animal Models for Parkinson's Disease Research: Trends in the 2000s. Int J Mol Sci 2019; 20:ijms20215402. [PMID: 31671557 PMCID: PMC6862023 DOI: 10.3390/ijms20215402] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is a chronic and progressive movement disorder and the second most common neurodegenerative disease. Although many studies have been conducted, there is an unmet clinical need to develop new treatments because, currently, only symptomatic therapies are available. To achieve this goal, clarification of the pathology is required. Attempts have been made to emulate human PD and various animal models have been developed over the decades. Neurotoxin models have been commonly used for PD research. Recently, advances in transgenic technology have enabled the development of genetic models that help to identify new approaches in PD research. However, PD animal model trends have not been investigated. Revealing the trends for PD research will be valuable for increasing our understanding of the positive and negative aspects of each model. In this article, we clarified the trends for animal models that were used to research PD in the 2000s, and we discussed each model based on these trends.
Collapse
|
31
|
Changes in motor behavior, neuropathology, and gut microbiota of a Batten disease mouse model following administration of acidified drinking water. Sci Rep 2019; 9:14962. [PMID: 31628420 PMCID: PMC6802212 DOI: 10.1038/s41598-019-51488-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022] Open
Abstract
CLN3 mutations cause the fatal neurodegenerative disorder, CLN3 Batten disease. The Cln3−/− mouse model displays characteristic features of the human disease including motor deficits. When mice received acidified drinking water (pH 2.5–2.9) instead of normal tap water (pH 8.4) for several generations, the motor skills of Cln3−/− mice normalized to control levels, indicating a disease-modifying effect of acidified water. Here we investigated if acidified water administered from postnatal day 21 has therapeutic benefits in Cln3−/− mice. Indeed, acidified water temporarily attenuated the motor deficits, had beneficial effects on behavioral parameters and prevented microglial activation in the brain of Cln3−/− mice. Interestingly, in control mice, acidified drinking water caused brain region-specific glial activation and significant changes in motor performance. Since the gut microbiota can influence neurological functions, we examined it in our disease model and found that the gut microbiota of Cln3−/− mice was markedly different from control mice, and acidified water differentially changed the gut microbiota composition in these mice. These results indicate that acidified water may provide therapeutic benefit to CLN3 Batten disease patients, and that the pH of drinking water is a major environmental factor that strongly influences the results of murine behavioral and pathological studies.
Collapse
|
32
|
Kwak SE, Cho SC, Bae JH, Lee J, Shin HE, Di Zhang D, Lee YI, Song W. Effects of exercise-induced apelin on muscle function and cognitive function in aged mice. Exp Gerontol 2019; 127:110710. [PMID: 31473200 DOI: 10.1016/j.exger.2019.110710] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/13/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Seong Eun Kwak
- Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Sung Chun Cho
- Well Aging Research Center, DGIST, Daegu, South Korea
| | - Jun Hyun Bae
- Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Jihyun Lee
- Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Hyung Eun Shin
- Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Di Di Zhang
- Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Yun-Il Lee
- Well Aging Research Center, DGIST, Daegu, South Korea; Department of New Biology, DGIST, Daegu, South Korea.
| | - Wook Song
- Institute of Sport Science, Seoul National University, Seoul, South Korea; Institue on Aging, Seoul National University, Seoul, South Korea.
| |
Collapse
|
33
|
A Parkinson's disease gene, DJ-1, regulates anti-inflammatory roles of astrocytes through prostaglandin D2 synthase expression. Neurobiol Dis 2019; 127:482-491. [DOI: 10.1016/j.nbd.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/07/2019] [Accepted: 04/03/2019] [Indexed: 01/24/2023] Open
|
34
|
Valek L, Auburger G, Tegeder I. Sensory neuropathy and nociception in rodent models of Parkinson's disease. Dis Model Mech 2019; 12:12/6/dmm039396. [PMID: 31248900 PMCID: PMC6602317 DOI: 10.1242/dmm.039396] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) often manifests with prodromal pain and sensory losses whose etiologies are not well understood. Multiple genetic and toxicity-based rodent models of PD partly recapitulate the histopathology and motor function deficits. Although far less studied, there is some evidence that rodents, similar to humans, develop sensory manifestations of the disease, which may precede motor disturbances and help to elucidate the underlying mechanisms of PD-associated pain at the molecular and neuron circuit levels. The present Review summarizes nociception and other sensory functions in frequently used rodent PD models within the context of the complex phenotypes. In terms of mechanisms, it appears that the acute loss of dopaminergic neurons in systemic toxicity models (MPTP, rotenone) primarily causes nociceptive hyperexcitability, presumably owing to a loss of inhibitory control, whereas genetic models primarily result in a progressive loss of heat perception, reflecting sensory fiber neuropathies. At the molecular level, neither α-synuclein deposits alone nor failure of mitophagy alone appear to be strong enough to result in axonal or synaptic pathology of nociceptive neurons that manifest at the behavioral level, and peripheral sensory loss may mask central ‘pain’ in behavioral tests. Hence, allostatic combinations or additional challenges and novel behavioral assessments are needed to better evaluate PD-associated sensory neuropathies and pain in rodents. Summary: Rodent models of Parkinson's disease partially develop prodromal somatosensory and olfactory dysfunctions reminiscent of sensory neuropathies in patients and reveal mechanistic insight, but data are incomplete and fragmented.
Collapse
Affiliation(s)
- Lucie Valek
- Institute of Clinical Pharmacology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Georg Auburger
- Experimental Neurology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, 60590 Frankfurt, Germany
| |
Collapse
|
35
|
Characterization of Motor and Non-Motor Behavioral Alterations in the Dj-1 (PARK7) Knockout Rat. J Mol Neurosci 2019; 69:298-311. [PMID: 31250274 DOI: 10.1007/s12031-019-01358-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/18/2019] [Indexed: 02/03/2023]
Abstract
Parkinson's disease is a neurodegenerative disorder that encompasses a constellation of motor and non-motor symptoms. The etiology of the disease is still poorly understood because of complex interactions between environmental and genetic risk factors. Using animal models to assess these risk factors may lead to a better understanding of disease manifestation. In this study, we assessed the Dj-1 knockout (KO) genetic rat model in a battery of motor and non-motor behaviors. We tested the Dj-1 KO rat, as well as age-matched wild-type (WT) control rats, in several sensorimotor tests at 2, 4, 7, and 13 months of age. The Dj-1-deficient rats were found to rear and groom less, and to have a shorter stride length than their WT counterparts, but to take more forelimb and hindlimb steps. In non-motor behavioral tasks, performed at several different ages, we evaluated the following: olfactory function, anxiety-like behavior, short-term memory, anhedonia, and stress coping behavior. Non-motor testing was conducted as early as 4.5 months and as late as 17 months of age. We found that Dj-1 KO animals displayed deficits in short-term spatial memory as early as 4.5 months of age during place preference testing, as well as impaired coping strategies in the forced swim test, which are consistent with a parkinsonian-like phenotype. In some instances, effects of chronic stress were evaluated in the Dj-1-deficient rats, as an initial test of an environmental challenge combined with a genetic disposition for PD. Although some of the results were mixed with differential effects across several of the behaviors, the combination of the changes we observed indicates that the Dj-1 KO rat may be a promising model for the assessment of the prodromal stage of Parkinson's disease, but further evaluation is necessary.
Collapse
|
36
|
Nasrolahi A, Safari F, Farhoudi M, Khosravi A, Farajdokht F, Bastaminejad S, Sandoghchian Shotorbani S, Mahmoudi J. Immune system and new avenues in Parkinson’s disease research and treatment. Rev Neurosci 2019; 30:709-727. [DOI: 10.1515/revneuro-2018-0105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder characterized by degeneration of dopaminergic neurons in the substantia nigra. However, although 200 years have now passed since the primary clinical description of PD by James Parkinson, the etiology and mechanisms of neuronal loss in this disease are still not fully understood. In addition to genetic and environmental factors, activation of immunologic responses seems to have a crucial role in PD pathology. Intraneuronal accumulation of α-synuclein (α-Syn), as the main pathological hallmark of PD, potentially mediates initiation of the autoimmune and inflammatory events through, possibly, auto-reactive T cells. While current therapeutic regimens are mainly used to symptomatically suppress PD signs, application of the disease-modifying therapies including immunomodulatory strategies may slow down the progressive neurodegeneration process of PD. The aim of this review is to summarize knowledge regarding previous studies on the relationships between autoimmune reactions and PD pathology as well as to discuss current opportunities for immunomodulatory therapy.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Fatemeh Safari
- Departmant of Medical Biotechnology, School of Advanced Medical Sciences and Technologies , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Afra Khosravi
- Department of Immunology, Faculty of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Saiyad Bastaminejad
- Department of Biochemistry and Molecular Medicine, School of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | | | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , P.O. 51666-14756, Tabriz , Iran , e-mail:
| |
Collapse
|
37
|
Gao L, Zhang Z, Xu W, Li T, Ying G, Qin B, Li J, Zheng J, Zhao T, Yan F, Zhu Y, Chen G. Natrium Benzoate Alleviates Neuronal Apoptosis via the DJ-1-Related Anti-oxidative Stress Pathway Involving Akt Phosphorylation in a Rat Model of Traumatic Spinal Cord Injury. Front Mol Neurosci 2019; 12:42. [PMID: 30853891 PMCID: PMC6395451 DOI: 10.3389/fnmol.2019.00042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022] Open
Abstract
This study aimed to explore the neuroprotective effects and mechanisms of natrium benzoate (NaB) and DJ-1 in attenuating reactive oxygen species (ROS)-induced neuronal apoptosis in traumatic spinal cord injury (t-SCI) in rats. T-SCI was induced by clip compression. The protein expression and neuronal apoptosis was evaluated by Western blotting, double immunofluorescence staining and transmission electron microscope (TEM). ROS level, spinal cord water content (SCWC) and Evans blue (EB) extravasation was also examined. Locomotor function was evaluated by Basso, Beattie, and Bresnahan (BBB) and inclined plane test (IPT) scores. We found that DJ-1 is expressed in spinal cord neurons and increased after t-SCI. At 24 h post-injury, the levels of DJ-1, p-Akt, SOD2, ROS, p-p38 MAPK/p38 MAPK ratio, and CC-3 increased, while the Bcl-2/Bax ratio decreased. NaB upregulated DJ-1, p-Akt, and SOD2, decreased ROS, p-p38 MAPK/p38 MAPK ratio, and CC-3, and increased the Bcl-2/Bax ratio, which were reversed by DJ-1 siRNA. The proportion of CC-3- and TUNEL-positive neurons also increased after t-SCI and was reduced by NaB. These effects were reversed by MK2206. Moreover, the level of oxDJ-1 increased after t-SCI, which was decreased by DJ-1 siRNA, NaB or the combination of them. NaB also reduced mitochondrial vacuolization, SCWC and EB extravasation, and improved locomotor function assessed by the BBB and IPT scores. In conclusion, NaB increased DJ-1, and thus reduced ROS and ROS-induced neuronal apoptosis by promoting Akt phosphorylation in t-SCI rats. NaB shows potential as a therapeutic agent for t-SCI, with DJ-1 as its main target.
Collapse
Affiliation(s)
- Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongyuan Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guangyu Ying
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Qin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingwei Zheng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tengfei Zhao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjian Zhu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Proteomic Profile of Carbonylated Proteins Screen Regulation of Apoptosis via CaMK Signaling in Response to Regular Aerobic Exercise. BIOMED RESEARCH INTERNATIONAL 2019; 2018:2828143. [PMID: 30662904 PMCID: PMC6312609 DOI: 10.1155/2018/2828143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/26/2018] [Indexed: 11/18/2022]
Abstract
To research carbonylated proteins and screen molecular targets in the rat striatum on regular aerobic exercise, male Sprague-Dawley rats (13 months old, n = 24) were randomly divided into middle-aged sedentary control (M-SED) and aerobic exercise (M-EX) groups (n = 12 each). Maximum oxygen consumption (VO2max) gradually increased from 50%-55% to 65%-70% for a total of 10 weeks. A total of 36 carbonylated proteins with modified oxidative sites were identified by Electrospray Ionization-Quadrupole-Time of Flight-Mass Spectrometer (ESI-Q-TOF-MS), including 17 carbonylated proteins unique to the M-SED group, calcium/calmodulin-dependent protein kinase type II subunit beta (CaMKIIβ), and heterogeneous nuclear ribonucleoprotein A2/B1 (Hnrnpa2b1), among others, and 19 specific to the M-EX group, ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCH-L1), and malic enzyme, among others. Regular aerobic exercise improved behavioral and stereological indicators, promoted normal apoptosis (P < 0.01), alleviated carbonylation of the CaMKIIβ and Hnrnpa2b1, but induced carbonylation of the UCH-L1, and significantly upregulated the expression levels of CaMKIIβ, CaMKIIα, and Vdac1 (p < 0.01) and Hnrnpa2b1 and UCH-L1 (p < 0.01), as well as the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin pathways (PI3K/Akt/mTOR) pathway-related genes Akt and mTOR. Regular aerobic exercise for 10 weeks (incremental for the first 6 weeks followed by constant loading for 4 weeks) enhanced carbonylation of CaMKIIβ, Hnrnpa2b1, and modulated apoptosis via activation of CaMK and phosphoinositide 3-kinase/protein kinase B/mTOR signaling. It also promoted normal apoptosis in the rat striatum, which may have protective effects in neurons.
Collapse
|
39
|
Faivre F, Joshi A, Bezard E, Barrot M. The hidden side of Parkinson’s disease: Studying pain, anxiety and depression in animal models. Neurosci Biobehav Rev 2019; 96:335-352. [DOI: 10.1016/j.neubiorev.2018.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/14/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022]
|
40
|
Marshall LJ, Willett C. Parkinson's disease research: adopting a more human perspective to accelerate advances. Drug Discov Today 2018; 23:1950-1961. [PMID: 30240875 DOI: 10.1016/j.drudis.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) affects 1% of the population over 60 years old and, with global increases in the aging population, presents huge economic and societal burdens. The etiology of PD remains unknown; most cases are idiopathic, presumed to result from genetic and environmental risk factors. Despite 200 years since the first description of PD, the mechanisms behind initiation and progression of the characteristic neurodegenerative processes are not known. Here, we review progress and limitations of the multiple PD animal models available and identify advances that could be implemented to better understand pathological processes, improve disease outcome, and reduce dependence on animal models. Lessons learned from reducing animal use in PD research could serve as guideposts for wider biomedical research.
Collapse
Affiliation(s)
- Lindsay J Marshall
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA
| | - Catherine Willett
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA.
| |
Collapse
|
41
|
Mita Y, Kataoka Y, Saito Y, Kashi T, Hayashi K, Iwasaki A, Imanishi T, Miyasaka T, Noguchi N. Distribution of oxidized DJ-1 in Parkinson's disease-related sites in the brain and in the peripheral tissues: effects of aging and a neurotoxin. Sci Rep 2018; 8:12056. [PMID: 30104666 PMCID: PMC6089991 DOI: 10.1038/s41598-018-30561-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023] Open
Abstract
DJ-1 plays an important role in antioxidant defenses, and a reactive cysteine at position 106 (Cys106) of DJ-1, a critical residue of its biological function, is oxidized under oxidative stress. DJ-1 oxidation has been reported in patients with Parkinson's disease (PD), but the relationship between DJ-1 oxidation and PD is still unclear. In the present study using specific antibody for Cys106-oxidized DJ-1 (oxDJ-1), we analyzed oxDJ-1 levels in the brain and peripheral tissues in young and aged mice and in a mouse model of PD induced using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). OxDJ-1 levels in the brain, heart, and skeletal muscle were high compared with other tissues. In the brain, oxDJ-1 was detected in PD-related brain sites such as the substantia nigra (SN) of the midbrain, olfactory bulb (OB), and striatum. In aged wild-type mice, oxDJ-1 levels in the OB, striatum, and heart tended to decrease, while those in the skeletal muscle increased significantly. Expression of dopamine-metabolizing enzymes significantly increased in the SN and OB of aged DJ-1-/- mice, accompanied by a complementary increase in glutathione peroxidase 1. MPTP treatment concordantly changed oxDJ-1 levels in PD-related brain sites and heart. These results indicate that the effects of physiological metabolism, aging, and neurotoxin change oxDJ-1 levels in PD-related brain sites, heart, and skeletal muscle where mitochondrial load is high, suggesting a substantial role of DJ-1 in antioxidant defenses and/or dopamine metabolism in these tissues.
Collapse
Affiliation(s)
- Yuichiro Mita
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yuto Kataoka
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yoshiro Saito
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan.
| | - Takuma Kashi
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Kojiro Hayashi
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Asa Iwasaki
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Takanori Imanishi
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Tomohiro Miyasaka
- Neuropathology, Department of Life and Medical Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Noriko Noguchi
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan.
| |
Collapse
|
42
|
|
43
|
Epigallocatechin-3-Gallate Protects and Prevents Paraquat-Induced Oxidative Stress and Neurodegeneration in Knockdown dj-1-β Drosophila melanogaster. Neurotox Res 2018; 34:401-416. [PMID: 29667128 DOI: 10.1007/s12640-018-9899-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/28/2022]
Abstract
Epigallocatechin-3-gallate (EGCG) is a polyhydroxyphenol constituent of green tea (e.g., Camellia sinensis) with known antioxidant properties. Due to these properties, others have proposed it as a potential therapeutic agent for the treatment of Parkinson's disease (PD). Previously, we demonstrated that EGCG prolonged the lifespan and locomotor activity in wild-type Canton-S flies exposed to the neurotoxicant paraquat (PQ), suggesting neuroprotective properties. Both gene mutations and environmental neurotoxicants (e.g., PQ) are factors involved in the development of PD. Thus, the first aim of this study was to create a suitable animal model of PD, which encompasses both of these factors. To create the model, we knocked down dj-1-β function specifically in the dopaminergic neurons to generate TH > dj-1-β-RNAi/+ Drosophila melanogaster flies. Next, we induced neurotoxicity in the transgenic flies with PQ. The second aim of this study was to validate the model by comparing the effects of vehicle, EGCG, and chemicals with known antioxidant and neuroprotective properties in vivo (e.g., propyl gallate and minocycline) on life-span, locomotor activity, lipid peroxidation, and neurodegeneration. The EGCG treatment provided protection and prevention from the PQ-induced reduction in the life-span and locomotor activity and from the PQ-induced increase in lipid peroxidation and neurodegeneration. These effects were augmented in the EGCG-treated flies when compared to the flies treated with either PG or MC. Altogether, these results suggest that the transgenic TH > dj-1-β-RNAi/+ flies treated with PQ serve as a suitable PD model for screening of potential therapeutic agents.
Collapse
|
44
|
Piston D, Alvarez-Erviti L, Bansal V, Gargano D, Yao Z, Szabadkai G, Odell M, Puno MR, Björkblom B, Maple-Grødem J, Breuer P, Kaut O, Larsen JP, Bonn S, Møller SG, Wüllner U, Schapira AHV, Gegg ME. DJ-1 is a redox sensitive adapter protein for high molecular weight complexes involved in regulation of catecholamine homeostasis. Hum Mol Genet 2018; 26:4028-4041. [PMID: 29016861 PMCID: PMC5886150 DOI: 10.1093/hmg/ddx294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/19/2017] [Indexed: 01/20/2023] Open
Abstract
DJ-1 is an oxidation sensitive protein encoded by the PARK7 gene. Mutations in PARK7 are a rare cause of familial recessive Parkinson’s disease (PD), but growing evidence suggests involvement of DJ-1 in idiopathic PD. The key clinical features of PD, rigidity and bradykinesia, result from neurotransmitter imbalance, particularly the catecholamines dopamine (DA) and noradrenaline. We report in human brain and human SH-SY5Y neuroblastoma cell lines that DJ-1 predominantly forms high molecular weight (HMW) complexes that included RNA metabolism proteins hnRNPA1 and PABP1 and the glycolysis enzyme GAPDH. In cell culture models the oxidation status of DJ-1 determined the specific complex composition. RNA sequencing indicated that oxidative changes to DJ-1 were concomitant with changes in mRNA transcripts mainly involved in catecholamine metabolism. Importantly, loss of DJ-1 function upon knock down (KD) or expression of the PD associated form L166P resulted in the absence of HMW DJ-1 complexes. In the KD model, the absence of DJ-1 complexes was accompanied by impairment in catecholamine homeostasis, with significant increases in intracellular DA and noraderenaline levels. These changes in catecholamines could be rescued by re-expression of DJ-1. This catecholamine imbalance may contribute to the particular vulnerability of dopaminergic and noradrenergic neurons to neurodegeneration in PARK7-related PD. Notably, oxidised DJ-1 was significantly decreased in idiopathic PD brain, suggesting altered complex function may also play a role in the more common sporadic form of the disease.
Collapse
Affiliation(s)
- Dominik Piston
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK.,Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Vikas Bansal
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Daniela Gargano
- Centre for Organelle Research, University of Stavanger, Stavanger, Norway
| | - Zhi Yao
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Mark Odell
- Department of Molecular and Applied Biosciences, University of Westminster, London, UK
| | - M Rhyan Puno
- Department of Molecular and Applied Biosciences, University of Westminster, London, UK
| | - Benny Björkblom
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Jodi Maple-Grødem
- Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Centre for Organelle Research, University of Stavanger, Stavanger, Norway
| | - Peter Breuer
- Department of Neurology, University of Bonn Medical Centre, Bonn, Germany
| | - Oliver Kaut
- Department of Neurology, University of Bonn Medical Centre, Bonn, Germany
| | - Jan Petter Larsen
- Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
| | - Stefan Bonn
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Simon Geir Møller
- Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Department of Biological Sciences, St. John's University, New York, NY, USA
| | - Ullrich Wüllner
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Matthew E Gegg
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK
| |
Collapse
|
45
|
Yang KM, Blue KV, Mulholland HM, Kurup MP, Kelm-Nelson CA, Ciucci MR. Characterization of oromotor and limb motor dysfunction in the DJ1 -/- model of Parkinson disease. Behav Brain Res 2018; 339:47-56. [PMID: 29109055 PMCID: PMC5729095 DOI: 10.1016/j.bbr.2017.10.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/28/2022]
Abstract
Parkinson disease (PD) is devastating to sensorimotor function that includes cranial/oromotor and limb motor deficits. However, the onset, progression, and neural correlates of PD-related dysfunctions are poorly understood. To address this gap, we used a genetic rat model of PD, DJ1 -/-, and hypothesized that motor deficits would manifest early in the disease process, be progressive in nature, and be related to pathologies in brainstem structures associated with sensorimotor function. The present study compares homozygous DJ1 -/- male rats to age-matched wild type controls. Progressive cranial sensorimotor function (ultrasonic vocalizations and tongue motor performance) and limb motor function (tapered balance beam) was analyzed at 2, 4, 6, and 8 months of age. Additionally, tyrosine hydroxylase cell counts were performed in the locus coeruleus and correlated to behavioral measures. We found that compared to wild type controls, DJ1 -/- show deficits in ultrasonic vocalizations as well as oromotor (tongue) deficits that were progressive. Overtime, DJ1 -/- rats cross a tapered balance beam with significantly decreased speed of traversal. Additionally, in the DJ1 -/-, tyrosine hydroxylase positive cells in the locus coeruleus are significantly reduced and are negatively correlated to oromotor behaviors. Characterizing the DJ1 -/- model of PD provides important foundational work necessary to define behavioral and early-onset biomarkers that parallels early-stage PD pathology in humans.
Collapse
Affiliation(s)
- Katie M Yang
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Katherine V Blue
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Haleigh M Mulholland
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Meghna P Kurup
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cynthia A Kelm-Nelson
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Michelle R Ciucci
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
46
|
Frøyset AK, Edson AJ, Gharbi N, Khan EA, Dondorp D, Bai Q, Tiraboschi E, Suster ML, Connolly JB, Burton EA, Fladmark KE. Astroglial DJ-1 over-expression up-regulates proteins involved in redox regulation and is neuroprotective in vivo. Redox Biol 2018. [PMID: 29525604 PMCID: PMC5854894 DOI: 10.1016/j.redox.2018.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
DJ-1, a Parkinson's disease-associated protein, is strongly up-regulated in reactive astrocytes in Parkinson's disease. This is proposed to represent a neuronal protective response, although the mechanism has not yet been identified. We have generated a transgenic zebrafish line with increased astroglial DJ-1 expression driven by regulatory elements from the zebrafish GFAP gene. Larvae from this transgenic line are protected from oxidative stress-induced injuries as caused by MPP+, a mitochondrial complex I inhibitor shown to induce dopaminergic cells death. In a global label-free proteomics analysis of wild type and transgenic larvae exposed to MPP+, 3418 proteins were identified, in which 366 proteins were differentially regulated. In particular, we identified enzymes belonging to primary metabolism to be among proteins affected by MPP+ in wild type animals, but not affected in the transgenic line. Moreover, by performing protein profiling on isolated astrocytes we showed that an increase in astrocytic DJ-1 expression up-regulated a large group of proteins associated with redox regulation, inflammation and mitochondrial respiration. The majority of these proteins have also been shown to be regulated by Nrf2. These findings provide a mechanistic insight into the protective role of astroglial up-regulation of DJ-1 and show that our transgenic zebrafish line with astrocytic DJ-1 over-expression can serve as a useful animal model to understand astrocyte-regulated neuroprotection associated with oxidative stress-related neurodegenerative disease. Increases astrocytic proteins linked to oxidative stress regulation & inflammation. Protects from MPP+-induced changes in central metabolism and protein nitrosylation. Protects from MPP+-induced tyrosine hydroxylase loss and motor deficits.
Collapse
Affiliation(s)
- Ann Kristin Frøyset
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway
| | - Amanda J Edson
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway
| | - Naouel Gharbi
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway
| | - Essa A Khan
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway
| | - Daniel Dondorp
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway
| | - Qing Bai
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ettore Tiraboschi
- Neural Circuits and Behaviour Group, Uni Research AS, Bergen N-5020, Norway
| | | | | | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kari E Fladmark
- Department of Biological Sciences, University of Bergen, Bergen N-5020, Norway.
| |
Collapse
|
47
|
Creed RB, Goldberg MS. New Developments in Genetic rat models of Parkinson's Disease. Mov Disord 2018; 33:717-729. [PMID: 29418019 DOI: 10.1002/mds.27296] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/04/2017] [Accepted: 12/10/2017] [Indexed: 12/12/2022] Open
Abstract
Preclinical research on Parkinson's disease has relied heavily on mouse and rat animal models. Initially, PD animal models were generated primarily by chemical neurotoxins that induce acute loss of dopaminergic neurons in the substantia nigra. On the discovery of genetic mutations causally linked to PD, mice were used more than rats to generate laboratory animals bearing PD-linked mutations because mutagenesis was more difficult in rats. Recent advances in technology for mammalian genome engineering and optimization of viral expression vectors have increased the use of genetic rat models of PD. Emerging research tools include "knockout" rats with disruption of genes in which mutations have been causally linked to PD, including LRRK2, α-synuclein, Parkin, PINK1, and DJ-1. Rats have also been increasingly used for transgenic and viral-mediated overexpression of genes relevant to PD, particularly α-synuclein. It may not be realistic to obtain a single animal model that completely reproduces every feature of a human disease as complex as PD. Nevertheless, compared with mice with the same mutations, many genetic rat animal models of PD better reproduce key aspects of PD including progressive loss of dopaminergic neurons in the substantia nigra, locomotor behavior deficits, and age-dependent formation of abnormal α-synuclein protein aggregates. Here we briefly review new developments in genetic rat models of PD that may have greater potential for identifying underlying mechanisms, for discovering novel therapeutic targets, and for developing greatly needed treatments to slow or halt disease progression. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rose B Creed
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Matthew S Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
48
|
DJ-1 deficiency impairs synaptic vesicle endocytosis and reavailability at nerve terminals. Proc Natl Acad Sci U S A 2018; 115:1629-1634. [PMID: 29386384 DOI: 10.1073/pnas.1708754115] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in DJ-1 (PARK7) are a known cause of early-onset autosomal recessive Parkinson's disease (PD). Accumulating evidence indicates that abnormalities of synaptic vesicle trafficking underlie the pathophysiological mechanism of PD. In the present study, we explored whether DJ-1 is involved in CNS synaptic function. DJ-1 deficiency impaired synaptic vesicle endocytosis and reavailability without inducing structural alterations in synapses. Familial mutants of DJ-1 (M26I, E64D, and L166P) were unable to rescue defective endocytosis of synaptic vesicles, whereas WT DJ-1 expression completely restored endocytic function in DJ-1 KO neurons. The defective synaptic endocytosis shown in DJ-1 KO neurons may be attributable to alterations in membrane cholesterol level. Thus, DJ-1 appears essential for synaptic vesicle endocytosis and reavailability, and impairment of this function by familial mutants of DJ-1 may be related to the pathogenesis of PD.
Collapse
|
49
|
Jiang P, Dickson DW. Parkinson's disease: experimental models and reality. Acta Neuropathol 2018; 135:13-32. [PMID: 29151169 PMCID: PMC5828522 DOI: 10.1007/s00401-017-1788-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a chronic, progressive movement disorder of adults and the second most common neurodegenerative disease after Alzheimer's disease. Neuropathologic diagnosis of PD requires moderate-to-marked neuronal loss in the ventrolateral substantia nigra pars compacta and α-synuclein (αS) Lewy body pathology. Nigrostriatal dopaminergic neurodegeneration correlates with the Parkinsonian motor features, but involvement of other peripheral and central nervous system regions leads to a wide range of non-motor features. Nigrostriatal dopaminergic neurodegeneration is shared with other parkinsonian disorders, including some genetic forms of parkinsonism, but many of these disorders do not have Lewy bodies. An ideal animal model for PD, therefore, should exhibit age-dependent and progressive dopaminergic neurodegeneration, motor dysfunction, and abnormal αS pathology. Rodent models of PD using genetic or toxin based strategies have been widely used in the past several decades to investigate the pathogenesis and therapeutics of PD, but few recapitulate all the major clinical and pathologic features of PD. It is likely that new strategies or better understanding of fundamental disease processes may facilitate development of better animal models. In this review, we highlight progress in generating rodent models of PD based on impairments of four major cellular functions: mitochondrial oxidative phosphorylation, autophagy-lysosomal metabolism, ubiquitin-proteasome protein degradation, and endoplasmic reticulum stress/unfolded protein response. We attempt to evaluate how impairment of these major cellular systems contribute to PD and how they can be exploited in rodent models. In addition, we review recent cell biological studies suggesting a link between αS aggregation and impairment of nuclear membrane integrity, as observed during cellular models of apoptosis. We also briefly discuss the role of incompetent phagocytic clearance and how this may be a factor to consider in developing new rodent models of PD.
Collapse
Affiliation(s)
- Peizhou Jiang
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Dennis W Dickson
- Neuropathology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
50
|
Choi DJ, Eun JH, Kim BG, Jou I, Park SM, Joe EH. A Parkinson's disease gene, DJ-1, repairs brain injury through Sox9 stabilization and astrogliosis. Glia 2017; 66:445-458. [PMID: 29105838 DOI: 10.1002/glia.23258] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022]
Abstract
Defects in repair of damaged brain accumulate injury and contribute to slow-developing neurodegeneration. Here, we report that a deficiency of DJ-1, a Parkinson's disease (PD) gene, delays repair of brain injury due to destabilization of Sox9, a positive regulator of astrogliosis. Stereotaxic injection of ATP into the brain striatum produces similar size of acute injury in wild-type and DJ-1-knockout (KO) mice. However, recovery of the injury is delayed in KO mice, which is confirmed by 9.4T magnetic resonance imaging and tyrosine hydroxylase immunostaining. DJ-1 regulates neurite outgrowth from damaged neurons in a non-cell autonomous manner. In DJ-1 KO brains and astrocytes, Sox9 protein levels are decreased due to enhanced ubiquitination, resulting in defects in astrogliosis and glial cell-derived neurotrophic factor/ brain-derived neurotrophic factor expression in injured brain and astrocytes. These results indicate that DJ-1 deficiency causes defects in astrocyte-mediated repair of brain damage, which may contribute to the development of PD.
Collapse
Affiliation(s)
- Dong-Joo Choi
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Jin-Hwa Eun
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Byung Gon Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Neurology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Ilo Jou
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Sang Myun Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| |
Collapse
|