1
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [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: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
2
|
Tassan Mazzocco M, Serra M, Maspero M, Coliva A, Presotto L, Casu MA, Morelli M, Moresco RM, Belloli S, Pinna A. Positive relation between dopamine neuron degeneration and metabolic connectivity disruption in the MPTP plus probenecid mouse model of Parkinson's disease. Exp Neurol 2024; 374:114704. [PMID: 38281587 DOI: 10.1016/j.expneurol.2024.114704] [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: 10/11/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
The clinical manifestation of Parkinson's disease (PD) appears when neurodegeneration is already advanced, compromising the efficacy of disease-modifying treatment approaches. Biomarkers to identify the early stages of PD are therefore of paramount importance for the advancement of the therapy of PD. In the present study, by using a mouse model of PD obtained by subchronic treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and the clearance inhibitor probenecid (MPTPp), we identified prodromal markers of PD by combining in vivo positron emission tomography (PET) imaging and ex vivo immunohistochemistry. Longitudinal PET imaging of the dopamine transporter (DAT) by [18F]-N-(3-fluoropropyl)-2β-carboxymethoxy-3β-(4-iodophenyl) nortropane ([18F]-FP-CIT), and brain glucose metabolism by 2-deoxy-2-[18F]-fluoroglucose ([18F]-FDG) were performed before MPTPp treatment and after 1, 3, and 10 MPTPp administrations, in order to assess relation between dopamine neuron integrity and brain connectivity. The results show that in vivo [18F]-FP-CIT in the dorsal striatum was not modified after the first administration of MPTPp, tended to decrease after 3 administrations, and significantly decreased after 10 MPTPp administrations. Post-mortem immunohistochemical analyses of DAT and tyrosine hydroxylase (TH) in the striatum showed a positive correlation with [18F]-FP-CIT, confirming the validity of repeated MPTPp-treated mice as a model that can reproduce the progressive pathological changes in the early phases of PD. Analysis of [18F]-FDG uptake in several brain areas connected to the striatum showed that metabolic connectivity was progressively disrupted, starting from the first MPTPp administration, and that significant connections between cortical and subcortical regions were lost after 10 MPTPp administrations, suggesting an association between dopamine neuron degeneration and connectivity disruption in this PD model. The results of this study provide a relevant model, where new drugs that can alleviate neurodegeneration in PD could be evaluated preclinically.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy; Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marco Maspero
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Luca Presotto
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; Department of Physics "G. Occhialini", University of Milano - Bicocca, Milan, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy; School of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy.
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| |
Collapse
|
3
|
Serra M, Di Maio A, Bassareo V, Nuzzo T, Errico F, Servillo F, Capasso M, Parekh P, Li Q, Thiolat ML, Bezard E, Calabresi P, Sulzer D, Carta M, Morelli M, Usiello A. Perturbation of serine enantiomers homeostasis in the striatum of MPTP-lesioned monkeys and mice reflects the extent of dopaminergic midbrain degeneration. Neurobiol Dis 2023; 184:106226. [PMID: 37451474 DOI: 10.1016/j.nbd.2023.106226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Loss of dopaminergic midbrain neurons perturbs l-serine and d-serine homeostasis in the post-mortem caudate putamen (CPu) of Parkinson's disease (PD) patients. However, it is unclear whether the severity of dopaminergic nigrostriatal degeneration plays a role in deregulating serine enantiomers' metabolism. Here, through high-performance liquid chromatography (HPLC), we measured the levels of these amino acids in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys and MPTP-plus-probenecid (MPTPp)-treated mice to determine whether and how dopaminergic midbrain degeneration affects the levels of serine enantiomers in various basal ganglia subregions. In addition, in the same brain regions, we measured the levels of key neuroactive amino acids modulating glutamatergic neurotransmission, including l-glutamate, glycine, l-aspartate, d-aspartate, and their precursors l-glutamine, l-asparagine. In monkeys, MPTP treatment produced severe denervation of nigrostriatal dopaminergic fibers (⁓75%) and increased the levels of serine enantiomers in the rostral putamen (rPut), but not in the subthalamic nucleus, and the lateral and medial portion of the globus pallidus. Moreover, this neurotoxin significantly reduced the protein expression of the astrocytic serine transporter ASCT1 and the glycolytic enzyme GAPDH in the rPut of monkeys. Conversely, concentrations of d-serine and l-serine, as well as ASCT1 and GAPDH expression were unaffected in the striatum of MPTPp-treated mice, which showed only mild dopaminergic degeneration (⁓30%). These findings unveil a link between the severity of dopaminergic nigrostriatal degeneration and striatal serine enantiomers concentration, ASCT1 and GAPDH expression. We hypothesize that the up-regulation of d-serine and l-serine levels occurs as a secondary response within a homeostatic loop to support the metabolic and neurotransmission demands imposed by the degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Anna Di Maio
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Tommaso Nuzzo
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesco Errico
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy
| | - Federica Servillo
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy
| | - Mario Capasso
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini, 5, Napoli 80131, Italy
| | - Pathik Parekh
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Qin Li
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Marie-Laure Thiolat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Erwan Bezard
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China; Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Paolo Calabresi
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy; Neurologia, Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Alessandro Usiello
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
4
|
Hyperbaric Oxygen Therapy Improves Parkinson’s Disease by Promoting Mitochondrial Biogenesis via the SIRT-1/PGC-1α Pathway. Biomolecules 2022; 12:biom12050661. [PMID: 35625589 PMCID: PMC9138219 DOI: 10.3390/biom12050661] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/25/2022] [Accepted: 04/28/2022] [Indexed: 12/30/2022] Open
Abstract
Hyperbaric oxygen therapy (HBOT) has been suggested as a potential adjunctive therapy for Parkinson’s disease (PD). PD is a neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The aim of this study was to investigate the protective mechanisms of HBOT on neurons and motor function in a 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and 1-methyl-4-phenylpyridinium (MPP+)-mediated neurotoxicity in SH-SY5Y cells on the potential protective capability. In vivo: male C57BL/6 mice were randomly divided into three groups: control, MPTP group and MPTP+HBOT group. The MPTP-treated mice were intraperitoneally received MPTP (20 mg/kg) four times at 2 h intervals within a day. The day after MPTP treatment, MPTP+HBOT mice were exposed to hyperbaric oxygen at 2.5 atmosphere absolute (ATA) with 100% oxygen for 1 h once daily for 7 consecutive days. In vitro: retinoic acid (RA)-differentiated SH-SY5Y cells were treated with MPP+ for 1 h followed by hyperbaric oxygen at 2.5 ATA with 100% oxygen for 1 h. The results showed that MPTP induced a significant loss in tyrosine hydroxylase (TH)-positive neurons in the SNpc of mice. HBOT treatment significantly increased the number of TH-positive neurons, with enhanced neurotrophic factor BDNF, decreased apoptotic signaling and attenuated inflammatory mediators in the midbrain of MPTP-treated mice. In addition, MPTP treatment decreased the locomotor activity and grip strength of mice, and these effects were shown to improve after HBOT treatment. Furthermore, MPTP decreased mitochondrial biogenesis signaling (SIRT-1, PGC-1α and TFAM), as well as mitochondrial marker VDAC expression, while HBOT treatment was shown to upregulate protein expression. In cell experiments, MPP+ reduced neurite length, while HBOT treatment attenuated neurite retraction. Conclusions: the effects of HBOT in MPTP-treated mice might come from promoting mitochondrial biogenesis, decreasing apoptotic signaling and attenuating inflammatory mediators in the midbrain, suggesting its potential benefits in PD treatment.
Collapse
|
5
|
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: 26] [Impact Index Per Article: 8.7] [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
|
6
|
Khairnar A, Ruda-Kucerova J, Arab A, Hadjistyllis C, Sejnoha Minsterova A, Shang Q, Chovsepian A, Drazanova E, Szabó N, Starcuk Z, Rektorova I, Pan-Montojo F. Diffusion kurtosis imaging detects the time-dependent progress of pathological changes in the oral rotenone mouse model of Parkinson's disease. J Neurochem 2021; 158:779-797. [PMID: 34107061 DOI: 10.1111/jnc.15449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 01/20/2023]
Abstract
Clinical diagnosis of Parkinson's disease (PD) occurs typically when a substantial proportion of dopaminergic neurons in the substantia nigra (SN) already died, and the first motor symptoms appear. Therefore, tools enabling the early diagnosis of PD are essential to identify early-stage PD patients in which neuroprotective treatments could have a significant impact. Here, we test the utility and sensitivity of the diffusion kurtosis imaging (DKI) in detecting progressive microstructural changes in several brain regions of mice exposed to chronic intragastric administration of rotenone, a mouse model that mimics the spatiotemporal progression of PD-like pathology from the ENS to the SN as described by Braak's staging. Our results show that DKI, especially kurtosis, can detect the progression of pathology-associated changes throughout the CNS. Increases in mean kurtosis were first observed in the dorsal motor nucleus of the vagus (DMV) after 2 months of exposure to rotenone and before the loss of dopaminergic neurons in the SN occurred. Remarkably, we also show that limited exposure to rotenone for 2 months is enough to trigger the progression of the disease in the absence of the environmental toxin, thus suggesting that once the first pathological changes in one region appear, they can self-perpetuate and progress within the CNS. Overall, our results show that DKI can be a useful radiological marker for the early detection and monitoring of PD pathology progression in patients with the potential to improve the clinical diagnosis and the development of neuroprotective treatments.
Collapse
Affiliation(s)
- Amit Khairnar
- Applied Neuroscience Research Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, India
| | - Jana Ruda-Kucerova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Anas Arab
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | - Alzbeta Sejnoha Minsterova
- Applied Neuroscience Research Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Qi Shang
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Alexandra Chovsepian
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Eva Drazanova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Nikoletta Szabó
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary.,Multi-modal and Functional Neuroimaging Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zenon Starcuk
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Irena Rektorova
- Applied Neuroscience Research Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Francisco Pan-Montojo
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Langley MR, Ghaisas S, Palanisamy BN, Ay M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Characterization of nonmotor behavioral impairments and their neurochemical mechanisms in the MitoPark mouse model of progressive neurodegeneration in Parkinson's disease. Exp Neurol 2021; 341:113716. [PMID: 33839143 PMCID: PMC9797183 DOI: 10.1016/j.expneurol.2021.113716] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/30/2022]
Abstract
Mitochondrial dysfunction has been implicated as a key player in the pathogenesis of Parkinson's disease (PD). The MitoPark mouse, a transgenic mitochondrial impairment model developed by specific inactivation of TFAM in dopaminergic neurons, spontaneously exhibits progressive motor deficits and neurodegeneration, recapitulating several features of PD. Since nonmotor symptoms are now recognized as important features of the prodromal stage of PD, we comprehensively assessed the clinically relevant motor and nonmotor deficiencies from ages 8-24 wk in both male and female MitoPark mice and their littermate controls. As expected, motor deficits in MitoPark mice began around 12-14 wk and became severe by 16-24 wk. Interestingly, MitoPark mice exhibited olfactory deficits in the novel and social scent tests as early as 10-12 wk as compared to age-matched littermate controls. Additionally, male MitoPark mice showed spatial memory deficits before female mice, beginning at 8 wk and becoming most severe at 16 wk, as determined by the Morris water maze. MitoPark mice between 16 and 24 wk spent more time immobile in forced swim and tail suspension tests, and made fewer entries into open arms of the elevated plus maze, indicating a depressive and anxiety-like phenotype, respectively. Importantly, depressive behavior as determined by immobility in forced swim test was reversible by antidepressant treatment with desipramine. Neurochemical and mechanistic studies revealed significant changes in CREB phosphorylation, BDNF, and catecholamine levels as well as neurogenesis in key brain regions. Collectively, our results indicate that MitoPark mice progressively exhibit deficits in olfactory discrimination, cognitive learning and memory, and anxiety- and depression-like behaviors as well as key neurochemical signaling associated with nonmotor deficits in PD. Thus, MitoPark mice can serve as an invaluable model for studying nonmotor deficits in addition to studying the motor deficits related to pathology in PD.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America.
| |
Collapse
|
8
|
Karuppagounder SS, Uthaythas S, Govindarajulu M, Ramesh S, Parameshwaran K, Dhanasekaran M. Caffeine, a natural methylxanthine nutraceutical, exerts dopaminergic neuroprotection. Neurochem Int 2021; 148:105066. [PMID: 34004240 DOI: 10.1016/j.neuint.2021.105066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 10/21/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that affects more than 10 million people worldwide. Oxidative stress and mitochondrial dysfunction play a significant role in altering the homeostasis of energy production and free radical generation. Current PD therapies are focused on reducing the cardinal symptoms rather than preventing disease progression in the patients. Adenosine A2A receptor (A2A R) antagonist (Istradephylline) combined with levodopa shows a promising therapy for PD. In animal studies, caffeine administration showed to improve motor functions and neuroprotective effect in the neurons. Caffeine is probably the most extensively used psychoactive substance. In this current study, we investigated the neuroprotective effect of caffeine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurodegeneration. Here, we demonstrate that caffeine improves behavioral and neurotransmitter recovery against MPTP-induced toxicity. Caffeine restores endogenous antioxidant levels and suppresses neuroinflammation. Our finding suggests that the blockage of A2AR is a promising disease-modifying therapy for PD. Target engagement strategies could be more beneficial in preventing disease progression rather than symptomatic reliefs in PD patients.
Collapse
Affiliation(s)
- Senthilkumar S Karuppagounder
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA.
| | - Subramaniam Uthaythas
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Sindhu Ramesh
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Koodeswaran Parameshwaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
9
|
Yang W, Hao W, Meng Z, Ding S, Li X, Zhang T, Huang W, Xu L, Zhang Y, Yang J, Gu X. Molecular Regulatory Mechanism and Toxicology of Neurodegenerative Processes in MPTP/Probenecid-Induced Progressive Parkinson's Disease Mice Model Revealed by Transcriptome. Mol Neurobiol 2021; 58:603-616. [PMID: 32997292 PMCID: PMC7843579 DOI: 10.1007/s12035-020-02128-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease caused by a variety of unclear complex pathogenic factors. The 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine/probenecid (MPTP/p)-induced progressive PD mice is a well-recognized classic model for studying PD, but the molecular toxicology of this model is still unclear. Here, for the first time, we report gradual neurodegenerative processes in MPTP/p-induced progressive PD mice model using RNA-seq. Transcriptional responses are orchestrated to regulate the expression of many genes in substantia nigra, such as Ntf3, Pitx3, Th, and Drd2, leading to the degeneration of dopaminergic neurons at last. We proposed that the established model could be divided into three phases based on their molecular toxicological features: "the stress response phase" which maintained the microenvironment homeostasis, "the pre-neurodegenerative phase" which demonstrated observed MPTP/p cytotoxicity and gradual degeneration of dopaminergic neurons, and "the neurodegenerative phase" which reflected distinct damage and dopaminergic neuron apoptotic process. Glia cells exhibited a certain protective effect on dopaminergic neurons in 3rd and 6th MPTP/p-induced cytotoxicity. But in 10th MPTP/p injection, glia cells play a promoting role in PD and tissue damages caused by oxidative stress. This study also indicated that the substantia nigra of PD mice showed unique patterns of changes at each stage. Moreover, neurotrophic signaling pathway, ECM-receptor interaction, oxidative phosphorylation, apoptosis and necroptosis were enriched at 3rd and 6th MPTP/p injection, which might be associated with the PD progress. This study provided an extensive data set of molecular toxicology for elucidating of PD progression and offered comprehensive theoretical knowledge for the development of new therapy.
Collapse
Affiliation(s)
- Weiwei Yang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenwen Hao
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhuo Meng
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyan Ding
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaodi Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tao Zhang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weixiao Huang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lian Xu
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yu Zhang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian Yang
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Xiaosong Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
10
|
Liu R, Li F, Zhao W. Long noncoding RNA NEAT1 knockdown inhibits MPP +-induced apoptosis, inflammation and cytotoxicity in SK-N-SH cells by regulating miR-212-5p/RAB3IP axis. Neurosci Lett 2020; 731:135060. [PMID: 32442477 DOI: 10.1016/j.neulet.2020.135060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Some long non-coding RNAs (lncRNAs) have been suggested to play critical roles in Parkinson's disease (PD) pathogenesis, including nuclear enriched abundant transcript 1 (NEAT1). The purpose of this study was to further elucidate the molecular mechanism of NEAT1 in PD. METHODS The expression levels of NEAT1, miR-212-5p and RAB3A-interacting protein (RAB3IP) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability and apoptosis were evaluated by Cell Counting Kit-8 (CCK-8) assay and flow cytometry analysis, respectively. Western blot analysis was applied to detect the protein expression of IL-1β, TNF-α and RAB3IP. The LDH activity, ROS generation and SOD activity were measured by Lactate LDH activity assay kit, ROS assay kit, and SOD activity assay kit, respectively. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to verify the relationship between miR-212-5p and NEAT1 or mRNA of RAB3IP. 1-methyl-4-phenylpyridinium ion (MPP+)-treated SK-N-SH cells were used as an in vitro model of PD. RESULTS NEAT1 and RAB3IP were upregulated while miR-212-5p was downregulated in SK-N-SH cells treated with MPP+. NEAT1 knockdown or miR-212-5p overexpression inhibited MPP+-induced apoptosis, inflammation and cytotoxicity in SK-N-SH cells. Moreover, miR-212-5p was a direct target of NEAT1 and its downregulation reversed the eff ;ects caused by NEAT1 knockdown in MPP+-induced SK-N-SH cells. Furthermore, RAB3IP was a downstream target of miR-212-5p and its overexpression attenuated the effects of miR-212-5p restoration in MPP+-induced SK-N-SH cells. Besides, NEAT1 acted as a molecular sponge of miR-212-5p to regulate RAB3IP expression. CONCLUSION NEAT1 knockdown suppressed MPP+-induced apoptosis, inflammation and cytotoxicity in SK-N-SH cells through regulating miR-212-5p and RAB3IP expression, providing a possible therapeutic strategy for PD patients.
Collapse
Affiliation(s)
- Ruiguang Liu
- Department of Neurology, Changle People's Hospital, Weifang, Shandong, China
| | - Fenlin Li
- Department of Geriatrics, Chengyang People's Hospital, Qingdao, Shandong, China
| | - Weijie Zhao
- Department of Geriatrics, Changle People's Hospital, Weifang, Shandong, China.
| |
Collapse
|
11
|
Advances in modelling alpha-synuclein-induced Parkinson’s diseases in rodents: Virus-based models versus inoculation of exogenous preformed toxic species. J Neurosci Methods 2020; 338:108685. [DOI: 10.1016/j.jneumeth.2020.108685] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 11/22/2022]
|
12
|
Wang F, Wang Z, Wang F, Dong K, Zhang J, Sun YJ, Liu CF, Xing MJ, Cheng X, Wei S, Zheng JW, Zhao XF, Wang XM, Fu J, Song HF. Comparative strategies for stem cell biodistribution in a preclinical study. Acta Pharmacol Sin 2020; 41:572-580. [PMID: 31705124 PMCID: PMC7470780 DOI: 10.1038/s41401-019-0313-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy represents the potential alternative effective strategy for some diseases that lack effective treatment currently. Correspondingly, it is crucial to establish high-sensitive and reliable quantification assay for tracing exogenous cell migration. In the present study, we first used both bioluminescence imaging (BLI) indirect labeling (human norepinephrine transporter-luciferase reporter system) and 89zirconium (89Zr)-hNSCs direct labeling combined with positron emission tomography/computer tomography (PET/CT) system for tracking human neural stem cells (hNSCs) migration into the brain via nasal administration in preclinical study. But the above two methods failed to give the biodistribution profile due to their low sensitivity. Considering its superior sensitivity and absolute quantitation capability, we developed and validated the droplet digital PCR (ddPCR) targeting species-specific gene in frozen and paraffin sections, slices, and whole blood with the sensitivity of 100–200 hNSCs. Accurate and high throughput quantification could be performed using ddPCR with the coefficient of variation (CVs) of lower quality control (LQC) below 30%. In combination with immunohistochemistry and ddPCR, we confirmed the migration of hNSCs into the brain via nasal administration, which supported the efficacy of hNSCs in MPTP-treated mice, an animal model of Parkinson’s disease. In conclusion, the present study is the first to report the application of ddPCR in the pharmacokinetics profile description of tracking of hNSCs in preclinical studies.
Collapse
|
13
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
14
|
Xu M, Bohlen JK, Moore C, Nipper MA, Finn DA, Jones CE, Lim MM, Meshul CK. Effects of sleep disruption on stress, nigrostriatal markers, and behavior in a chronic/progressive MPTP male mouse model of parkinsonism. J Neurosci Res 2019; 97:1706-1719. [PMID: 31535395 PMCID: PMC6801095 DOI: 10.1002/jnr.24520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/14/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Sleep complaints are an early clinical symptom of neurodegenerative disorders. Patients with Parkinson's disease (PD) experience sleep disruption (SD). The objective of this study was to determine if preexisting, chronic SD leads to a greater loss of tyrosine hydroxylase (TH) within the striatum and the substantia nigra following chronic/progressive exposure with the neurotoxin, 1-methyl-2-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Male mice underwent chronic SD for 4 weeks, then injected with vehicle (VEH) or increasing doses of MPTP for 4 weeks. There was a significant decrease in the plasma corticosterone levels in the MPTP group, an increase in the SD group, and a return to the VEH levels in the SD+MPTP group. Protein expression levels for TH in the striatum (terminals) and substantia nigra pars compacta (dopamine [DA] cell counts) revealed up to a 78% and 38% decrease, respectively, in the MPTP and SD+MPTP groups compared to their relevant VEH and SD groups. DA transporter protein expression increased in the striatum in the MPTP versus VEH group and in the SN/midbrain between the SD+MPTP and the VEH group. There was a main effect of MPTP on various gait measures (e.g., braking) relative to the SD or VEH groups. In the SD+MPTP group, there were no differences compared to the VEH group. Thus, SD, prior to administration of MPTP, has effects on serum corticosterone and gait but more importantly does not potentiate greater loss of TH within the nigrostriatal pathway compared to the MPTP group, suggesting that in PD patients with SD, there is no exacerbation of the DA cell loss.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Corpus Striatum/enzymology
- Corpus Striatum/pathology
- Corticosterone/blood
- Disease Models, Animal
- Dopamine Plasma Membrane Transport Proteins/analysis
- Gait Disorders, Neurologic/etiology
- Gait Disorders, Neurologic/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Nerve Tissue Proteins/analysis
- Oxidopamine/toxicity
- Parkinsonian Disorders/complications
- Parkinsonian Disorders/metabolism
- Single-Blind Method
- Sleep Disorders, Intrinsic/blood
- Sleep Disorders, Intrinsic/etiology
- Sleep Disorders, Intrinsic/physiopathology
- Stress, Physiological
- Substantia Nigra/enzymology
- Substantia Nigra/pathology
- Tyrosine 3-Monooxygenase/analysis
- Vesicular Monoamine Transport Proteins/analysis
Collapse
Affiliation(s)
- Mo Xu
- Research Services, VA Medical Center/Portland, OR
| | | | | | | | - Deborah A. Finn
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
| | - Carolyn E. Jones
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
| | - Miranda M. Lim
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
- Department of Neurology, Oregon Health & Science University
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University
| | - Charles K. Meshul
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
- Department of Pathology, Oregon Health & Science University
| |
Collapse
|
15
|
Arab A, Ruda-Kucerova J, Minsterova A, Drazanova E, Szabó N, Starcuk Z, Rektorova I, Khairnar A. Diffusion Kurtosis Imaging Detects Microstructural Changes in a Methamphetamine-Induced Mouse Model of Parkinson’s Disease. Neurotox Res 2019; 36:724-735. [DOI: 10.1007/s12640-019-00068-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022]
|
16
|
Fuzzati-Armentero MT, Cerri S, Blandini F. Peripheral-Central Neuroimmune Crosstalk in Parkinson's Disease: What Do Patients and Animal Models Tell Us? Front Neurol 2019; 10:232. [PMID: 30941089 PMCID: PMC6433876 DOI: 10.3389/fneur.2019.00232] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is no longer considered an immune privileged organ and neuroinflammation has long been associated with Parkinson's disease. Accumulating evidence demonstrates that innate and adaptive responses take place in the CNS. The extent to which peripheral immune alterations impacts on the CNS, or vice and versa, is, however, still a matter of debate. Gaining a better knowledge of the molecular and cellular immune dysfunctions present in these two compartments and clarifying their mutual interactions is a fundamental step in understanding and preventing Parkinson's disease (PD) pathogenesis. This review provides an overview of the current knowledge on inflammatory processes evidenced both in PD patients and in toxin-induced animal models of the disease. It discusses differences and similarities between human and animal studies in the context of neuroinflammation and immune responses and how they have guided therapeutic strategies to slow down disease progression. Future longitudinal studies are necessary and can help gain a better understanding on peripheral-central nervous system crosstalk to improve therapeutic strategies for PD.
Collapse
|
17
|
Bhurtel S, Katila N, Srivastav S, Neupane S, Choi DY. Mechanistic comparison between MPTP and rotenone neurotoxicity in mice. Neurotoxicology 2019; 71:113-121. [DOI: 10.1016/j.neuro.2018.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/24/2018] [Accepted: 12/28/2018] [Indexed: 01/02/2023]
|
18
|
Lecca D, Janda E, Mulas G, Diana A, Martino C, Angius F, Spolitu S, Casu MA, Simbula G, Boi L, Batetta B, Spiga S, Carta AR. Boosting phagocytosis and anti-inflammatory phenotype in microglia mediates neuroprotection by PPARγ agonist MDG548 in Parkinson's disease models. Br J Pharmacol 2018; 175:3298-3314. [PMID: 29570770 DOI: 10.1111/bph.14214] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 03/02/2018] [Accepted: 03/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Microglial phenotype and phagocytic activity are deregulated in Parkinson's disease (PD). PPARγ agonists are neuroprotective in experimental PD, but their role in regulating microglial phenotype and phagocytosis has been poorly investigated. We addressed it by using the PPARγ agonist MDG548. EXPERIMENTAL APPROACH Murine microglial cell line MMGT12 was stimulated with LPS and/or MDG548, and their effect on phagocytosis of fluorescent microspheres or necrotic neurons was investigated by flow cytometry. Cytokines and markers of microglia phenotype, such as mannose receptor C type 1; MRC1), Ym1 and CD68 were measured by elisa and fluorescent immunohistochemistry. Levels of Beclin-1, which plays a role in microglial phagocytosis, were measured by Western blotting. In the in vivo MPTP-probenecid (MPTPp) model of PD in mice, MDG548 was tested on motor impairment, nigral neurodegeneration, microglial activation and phenotype. KEY RESULTS In LPS-stimulated microglia, MDG548 increased phagocytosis of both latex beads and necrotic cells, up-regulated the expression of MRC1, CD68 and to a lesser extent IL-10, while blocking the LPS-induced increase of TNF-α and iNOS. MDG548 also induced Beclin-1. Chronic MPTPp treatment in mice down-regulated MRC1 and TGF-β and up-regulated TNF-α and IL-1β immunoreactivity in activated CD11b-positive microglia, causing the death of nigral dopaminergic neurons. MDG548 arrested MPTPp-induced cell death, enhanced MRC1 and restored cytokine levels. CONCLUSIONS AND IMPLICATIONS This study adds a novel mechanism for PPARγ-mediated neuroprotection in PD and suggests that increasing phagocytic activity and anti-inflammatory markers may represent an effective disease-modifying approach.
Collapse
Affiliation(s)
- Daniela Lecca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Giovanna Mulas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Concetta Martino
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Spolitu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Gabriella Simbula
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Barbara Batetta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
19
|
Vingill S, Connor-Robson N, Wade-Martins R. Are rodent models of Parkinson's disease behaving as they should? Behav Brain Res 2017; 352:133-141. [PMID: 29074404 DOI: 10.1016/j.bbr.2017.10.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 12/20/2022]
Abstract
In recent years our understanding of Parkinson's disease has expanded both in terms of pathological hallmarks as well as relevant genetic influences. In parallel with the aetiological discoveries a multitude of PD animal models have been established. The vast majority of these are rodent models based on environmental, genetic and mechanistic insight. A major challenge in many of these models is their ability to only recapitulate some of the complex disease features seen in humans. Although symptom alleviation and clinical signs are of utmost importance in therapeutic research many of these models lack comprehensive behavioural testing. While non-motor symptoms become increasingly important as early diagnostic markers in PD, they are poorly characterized in rodents. In this review we look at well-established and more recent animal models of PD in terms of behavioural characterization and discuss how they can best contribute to progression in Parkinson's research.
Collapse
Affiliation(s)
- Siv Vingill
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK.
| | - Natalie Connor-Robson
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
20
|
Katila N, Bhurtel S, Shadfar S, Srivastav S, Neupane S, Ojha U, Jeong GS, Choi DY. Metformin lowers α-synuclein phosphorylation and upregulates neurotrophic factor in the MPTP mouse model of Parkinson's disease. Neuropharmacology 2017; 125:396-407. [PMID: 28807678 DOI: 10.1016/j.neuropharm.2017.08.015] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/01/2017] [Accepted: 08/10/2017] [Indexed: 10/19/2022]
Abstract
In spite of the massive research for the identification of neurorestorative or neuroprotective intervention for curing Parkinson's disease (PD), there is still lack of clinically proven neuroprotective agents. Metformin, a common anti-hyperglycemic drug has been known to possess neuroprotective properties. However, specific mechanisms by which metformin protects neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity remain to be elucidated. In this study, we assessed the neuroprotective effects of metformin in the subchronic MPTP model of PD, and explored its feasible mechanisms for neuroprotection. Animals received saline or MPTP injection (30 mg/kg/day) for the first 7 days, and then saline or metformin (200 mg/kg/day) for the next 7 days. Immunohistochemical stainings showed that metformin rescued the tyrosine hydroxylase-positive neurons and attenuated astroglial activation in the nigrostriatal pathway. In parallel, metformin restored dopamine depletion and behavioral impairments exerted by MPTP. Western blot analysis revealed that metformin ameliorated MPTP-induced α-synuclein phosphorylation which was accompanied by increased methylation of protein phosphatase 2A (PP2A), a phosphatase related to α-synuclein dephosphorylation. Moreover, the metformin regimen significantly increased the level of brain derived neurotrophic factor in the substantia nigra, and activated signaling pathways related to cell survival. Proof of concept study revealed that inhibition of PP2A or tropomyosin receptor kinase B reversed neuroprotective property of metformin in SH-SY5Y cells. Our results indicate that metformin provides neuroprotection against MPTP neurotoxicity, which might be mediated by inhibition of α-synuclein phosphorylation and induction of neurotrophic factors.
Collapse
Affiliation(s)
- Nikita Katila
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sunil Bhurtel
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sina Shadfar
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sunil Srivastav
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sabita Neupane
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Uttam Ojha
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Gil-Saeng Jeong
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Daegu 42601, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
21
|
Contrasting effects of selective MAGL and FAAH inhibition on dopamine depletion and GDNF expression in a chronic MPTP mouse model of Parkinson's disease. Neurochem Int 2017; 110:14-24. [PMID: 28826718 DOI: 10.1016/j.neuint.2017.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 06/08/2017] [Accepted: 08/03/2017] [Indexed: 01/07/2023]
Abstract
The modulation of the brain endocannabinoid system has been identified as an option to treat neurodegenerative diseases including Parkinson's disease (PD). Especially the elevation of endocannabinoid levels by inhibition of hydrolytic degradation represents a valuable approach. To evaluate whether monoacylglycerol lipase (MAGL) or fatty acid amide hydrolase (FAAH) inhibition could be beneficial for PD, we examined in parallel the therapeutic potential of the highly selective MAGL inhibitor KML29 elevating 2-arachidonoylglyerol (2-AG) levels and the highly selective FAAH inhibitor PF-3845 elevating anandamide (AEA) levels in a chronic methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/probenecid) mouse model of PD. Chronic administration of KML29 (10 mg/kg) but not PF-3845 (10 mg/kg) attenuated striatal MPTP/probenecid-induced dopamine depletion. Furthermore, KML29 induced an increase in Gdnf but not Bdnf expression, whereas PF-3845 decreased the MPTP/probenecid-induced Cnr2 expression without any effects on neurotrophin expression. Investigation of treatment-naïve striatal mRNA levels revealed a high presence of Gdnf and Mgll in contrast to Bdnf and Faah. Treatment of primary mouse microglia with 2-AG increased Gdnf but not Bdnf expression, suggesting that microglia might mediate the observed KML29-induced increase in Gdnf. In summary, pharmacological MAGL but not FAAH inhibition in the chronic MPTP/probenecid model attenuated the MPTP/probenecid-induced effects on striatal dopamine levels which were accompanied by an increase in 2-AG levels.
Collapse
|
22
|
Joers V, Tansey MG, Mulas G, Carta AR. Microglial phenotypes in Parkinson's disease and animal models of the disease. Prog Neurobiol 2017; 155:57-75. [PMID: 27107797 PMCID: PMC5073045 DOI: 10.1016/j.pneurobio.2016.04.006] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/19/2022]
Abstract
Over the last decade the important concept has emerged that microglia, similar to other tissue macrophages, assume different phenotypes and serve several effector functions, generating the theory that activated microglia can be organized by their pro-inflammatory or anti-inflammatory and repairing functions. Importantly, microglia exist in a heterogenous population and their phenotypes are not permanently polarized into two categories; they exist along a continuum where they acquire different profiles based on their local environment. In Parkinson's disease (PD), neuroinflammation and microglia activation are considered neuropathological hallmarks, however their precise role in relation to disease progression is not clear, yet represent a critical challenge in the search of disease-modifying strategies. This review will critically address current knowledge on the activation states of microglia as well as microglial phenotypes found in PD and in animal models of PD, focusing on the expression of surface molecules as well as pro-inflammatory and anti-inflammatory cytokine production during the disease process. While human studies have reported an elevation of both pro- or anti-inflammatory markers in the serum and CSF of PD patients, animal models have provided insights on dynamic changes of microglia phenotypes in relation to disease progression especially prior to the development of motor deficits. We also review recent evidence of malfunction at multiple steps of NFκB signaling that may have a causal interrelationship with pathological microglia activation in animal models of PD. Finally, we discuss the immune-modifying strategies that have been explored regarding mechanisms of chronic microglial activation.
Collapse
Affiliation(s)
- Valerie Joers
- Department of Physiology, Emory University, Atlanta, GA, United States; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Malú G Tansey
- Department of Physiology, Emory University, Atlanta, GA, United States.
| | - Giovanna Mulas
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Italy.
| |
Collapse
|
23
|
Viveros-Paredes JM, González-Castañeda RE, Gertsch J, Chaparro-Huerta V, López-Roa RI, Vázquez-Valls E, Beas-Zarate C, Camins-Espuny A, Flores-Soto ME. Neuroprotective Effects of β-Caryophyllene against Dopaminergic Neuron Injury in a Murine Model of Parkinson's Disease Induced by MPTP. Pharmaceuticals (Basel) 2017; 10:E60. [PMID: 28684694 PMCID: PMC5620604 DOI: 10.3390/ph10030060] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders and is characterized by the loss of dopaminergic neurons in the substantia nigra (SN). Although the causes of PD are not understood, evidence suggests that its pathogenesis is associated with oxidative stress and inflammation. Recent studies have suggested a protective role of the cannabinoid signalling system in PD. β-caryophyllene (BCP) is a natural bicyclic sesquiterpene that is an agonist of the cannabinoid type 2 receptor (CB2R). Previous studies have suggested that BCP exerts prophylactic and/or curative effects against inflammatory bowel disease through its antioxidative and/or anti-inflammatory action. The present study describes the neuroprotective effects of BCP in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced murine model of PD, and we report the results of our investigation of its neuroprotective mechanism in neurons and glial cells. In the murine model, BCP pretreatment ameliorated motor dysfunction, protected against dopaminergic neuronal losses in the SN and striatum, and alleviated MPTP-induced glia activation. Additionally, BCP inhibited the levels of inflammatory cytokines in the nigrostriatal system. The observed neuroprotection and inhibited glia activation were reversed upon treatment with the CB2R selective antagonist AM630, confirming the involvement of the CB2R. These results indicate that BCP acts via multiple neuroprotective mechanisms in our murine model and suggest that BCP may be viewed as a potential treatment and/or preventative agent for PD.
Collapse
Affiliation(s)
- Juan M Viveros-Paredes
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
| | - Rocio E González-Castañeda
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| | - Juerg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR Trans Cure, University of Bern, CH-3012 Bern, Switzerland.
| | - Veronica Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, 44421 Guadalajara, Mexico.
| | - Rocio I López-Roa
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
| | - Eduardo Vázquez-Valls
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, 44421 Guadalajara, Mexico.
| | - Carlos Beas-Zarate
- Laboratorio de Regeneración y Desarrollo Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| | - Antoni Camins-Espuny
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciencias de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain.
| | - Mario E Flores-Soto
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| |
Collapse
|
24
|
Carelli S, Giallongo T, Viaggi C, Latorre E, Gombalova Z, Raspa A, Mazza M, Vaglini F, Di Giulio AM, Gorio A. Recovery from experimental parkinsonism by intrastriatal application of erythropoietin or EPO-releasing neural precursors. Neuropharmacology 2017; 119:76-90. [PMID: 28373075 DOI: 10.1016/j.neuropharm.2017.03.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/15/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
Abstract
An extensive literature has shown a powerful neuroprotective action of Erythropoietin (EPO) both in vivo and in vitro. This study shows that EPO, whether ectopically administered or released by neural precursors, does reverse MPTP-induced parkinsonism in mice. Unilateral stereotaxic injection of 2.5 × 105 erythropoietin-releasing neural precursor cells (Er-NPCs) rescued degenerating striatal dopaminergic neurons and promoted behavioral recovery as shown by three independent behavioral tests. These effects were replicated through direct intrastriatal administration of recombinant human EPO. At the end of the observational period, most of the transplanted Er-NPCs were vital and migrated via the striatum to reach Substantia Nigra. The restorative effects appear to be mediated by EPO since co-injection of anti-EPO or anti-EPOR antibodies antagonized the positive outcomes. Furthermore, this report supports the neuroprotective action of EPO, which may also be achieved via administration of EPO-releasing cells such as Er-NPCs.
Collapse
Affiliation(s)
- Stephana Carelli
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy.
| | - Toniella Giallongo
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Cristina Viaggi
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Italy
| | - Elisa Latorre
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Zuzana Gombalova
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Andrea Raspa
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Massimiliano Mazza
- Experimental Oncology Department, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Francesca Vaglini
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Italy
| | - Anna Maria Di Giulio
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Alfredo Gorio
- Laboratories of Pharmacology, Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
25
|
Early and progressive microstructural brain changes in mice overexpressing human α-Synuclein detected by diffusion kurtosis imaging. Brain Behav Immun 2017; 61:197-208. [PMID: 27923670 DOI: 10.1016/j.bbi.2016.11.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/18/2016] [Accepted: 11/27/2016] [Indexed: 12/27/2022] Open
Abstract
Diffusion kurtosis imaging (DKI) is sensitive in detecting α-Synuclein (α-Syn) accumulation-associated microstructural changes at late stages of the pathology in α-Syn overexpressing TNWT-61 mice. The aim of this study was to perform DKI in young TNWT-61 mice when α-Syn starts to accumulate and to compare the imaging results with an analysis of motor and memory impairment and α-Syn levels. Three-month-old (3mo) and six-month-old (6mo) mice underwent DKI scanning using the Bruker Avance 9.4T magnetic resonance imaging system. Region of interest (ROI) analyses were performed in the gray matter; tract-based spatial statistics (TBSS) analyses were performed in the white matter. In the same mice, α-Syn expression was evaluated using quantitative immunofluorescence. Mean kurtosis (MK) was the best differentiator between TNWT-61 mice and wildtype (WT) mice. We found increases in MK in 3mo TNWT-61 mice in the striatum and thalamus but not in the substantia nigra (SN), hippocampus, or sensorimotor cortex, even though the immunoreactivity of human α-Syn was similar or even higher in the latter regions. Increases in MK in the SN were detected in 6mo mice. These findings indicate that α-Syn accumulation-associated changes may start in areas with a high density of dopaminergic nerve terminals. We also found TBSS changes in white matter only at 6mo, suggesting α-Syn accumulation-associated changes start in the gray matter and later progress to the white matter. MK is able to detect microstructural changes induced by α-Syn overexpression in TNWT-61 mice and could be a useful clinical tool for detecting early-stage Parkinson's disease in human patients.
Collapse
|
26
|
Gu PS, Moon M, Choi JG, Oh MS. Mulberry fruit ameliorates Parkinson's-disease-related pathology by reducing α-synuclein and ubiquitin levels in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid model. J Nutr Biochem 2017; 39:15-21. [DOI: 10.1016/j.jnutbio.2016.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 08/03/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
27
|
Selenotranscriptomic Analyses Identify Signature Selenoproteins in Brain Regions in a Mouse Model of Parkinson's Disease. PLoS One 2016; 11:e0163372. [PMID: 27656880 PMCID: PMC5033372 DOI: 10.1371/journal.pone.0163372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 01/22/2023] Open
Abstract
Genes of selenoproteome have been increasingly implicated in various aspects of neurobiology and neurological disorders, but remain largely elusive in Parkinson’s disease (PD). In this study, we investigated the selenotranscriptome (24 selenoproteins in total) in five brain regions (cerebellum, substantia nigra, cortex, pons and hippocampus) by real time qPCR in a two-phase manner using a mouse model of chronic PD. A wide range of changes in selenotranscriptome was observed in a manner depending on selenoproteins and brain regions. While Selv mRNA was not detectable and Dio1& 3 mRNA levels were not affected, 1, 11 and 9 selenoproteins displayed patterns of increase only, decrease only, and mixed response, respectively, in these brain regions of PD mice. In particular, the mRNA expression of Gpx1-4 showed only a decreased trend in the PD mouse brains. In substantia nigra, levels of 17 selenoprotein mRNAs were significantly decreased whereas no selenoprotein was up-regulated in the PD mice. In contrast, the majority of selenotranscriptome did not change and a few selenoprotein mRNAs that respond displayed a mixed pattern of up- and down-regulation in cerebellum, cortex, hippocampus, and/or pons of the PD mice. Gpx4, Sep15, Selm, Sepw1, and Sepp1 mRNAs were most abundant across all these five brain regions. Our results showed differential responses of selenoproteins in various brain regions of the PD mouse model, providing critical selenotranscriptomic profiling for future functional investigation of individual selenoprotein in PD etiology.
Collapse
|
28
|
Succinobucol, a Non-Statin Hypocholesterolemic Drug, Prevents Premotor Symptoms and Nigrostriatal Neurodegeneration in an Experimental Model of Parkinson's Disease. Mol Neurobiol 2016; 54:1513-1530. [PMID: 26852411 DOI: 10.1007/s12035-016-9747-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/26/2016] [Indexed: 01/04/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by non-motor and motor disabilities. This study investigated whether succinobucol (SUC) could mitigate nigrostriatal injury caused by intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in mice. Moreover, the effects of SUC against MPTP-induced behavioral impairments and neurochemical changes were also evaluated. The quantification of tyrosine hydroxylase-positive (TH+) cells was also performed in primary mesencephalic cultures to evaluate the effects of SUC against 1-methyl-4-phenylpyridinium (MPP+) toxicity in vitro. C57BL/6 mice were treated with SUC (10 mg/kg/day, intragastric (i.g.)) for 30 days, and thereafter, animals received MPTP infusion (1 mg/nostril) and SUC treatment continued for additional 15 days. MPTP-infused animals displayed significant non-motor symptoms including olfactory and short-term memory deficits evaluated in the olfactory discrimination, social recognition, and water maze tasks. These behavioral impairments were accompanied by inhibition of mitochondrial NADH dehydrogenase activity (complex I), as well as significant decrease of TH and dopamine transporter (DAT) immunoreactivity in the substantia nigra pars compacta and striatum. Although SUC treatment did not rescue NADH dehydrogenase activity inhibition, it was able to blunt MPTP-induced behavioral impairments and prevented the decrease in TH and DAT immunoreactivities in substantia nigra (SN) and striatum. SUC also suppressed striatal astroglial activation and increased interleukin-6 levels in MPTP-intoxicated mice. Furthermore, SUC significantly prevented the loss of TH+ neurons induced by MPP+ in primary mesencephalic cultures. These results provide new evidence that SUC treatment counteracts early non-motor symptoms and neurodegeneration/neuroinflammation in the nigrostriatal pathway induced by intranasal MPTP administration in mice by modulating events downstream to the mitochondrial NADH dehydrogenase inhibition.
Collapse
|
29
|
Baranyi M, Porceddu PF, Gölöncsér F, Kulcsár S, Otrokocsi L, Kittel Á, Pinna A, Frau L, Huleatt PB, Khoo ML, Chai CLL, Dunkel P, Mátyus P, Morelli M, Sperlágh B. Novel (Hetero)arylalkenyl propargylamine compounds are protective in toxin-induced models of Parkinson's disease. Mol Neurodegener 2016; 11:6. [PMID: 26758813 PMCID: PMC4711075 DOI: 10.1186/s13024-015-0067-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/22/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction, oxidative stress and their interplay are core pathological features of Parkinson's disease. In dopaminergic neurons, monoamines and their metabolites provide an additional source of reactive free radicals during their breakdown by monoamine oxidase or auto-oxidation. Moreover, mitochondrial dysfunction and oxidative stress have a supraadditive impact on the pathological, cytoplasmic accumulation of dopamine and its subsequent release. Here we report the effects of a novel series of potent and selective MAO-B inhibitory (hetero)arylalkenylpropargylamine compounds having protective properties against the supraadditive effect of mitochondrial dysfunction and oxidative stress. RESULTS The (hetero)arylalkenylpropargylamines were tested in vitro, on acute rat striatal slices, pretreated with the complex I inhibitor rotenone and in vivo, using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced acute, subchronic, and chronic experimental models of Parkinson's disease in mice. The compounds exhibited consistent protective effects against i) in vitro oxidative stress induced pathological dopamine release and the formation of toxic dopamine quinone in the rat striatum and rescued tyrosine hydroxylase positive neurons in the substantia nigra after rotenone treatment; ii) in vivo MPTP-induced striatal dopamine depletion and motor dysfunction in mice using acute and subchronic, delayed application protocols. One compound (SZV558) was also examined and proved to be protective in a chronic mouse model of MPTP plus probenecid (MPTPp) administration, which induces a progressive loss of nigrostriatal dopaminergic neurons. CONCLUSIONS Simultaneous inhibition of MAO-B and oxidative stress induced pathological dopamine release by the novel propargylamines is protective in animal models and seems a plausible strategy to combat Parkinson's disease.
Collapse
Affiliation(s)
- Mária Baranyi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy.
| | - Flóra Gölöncsér
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary. .,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary.
| | - Szabina Kulcsár
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Lilla Otrokocsi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary. .,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary.
| | - Ágnes Kittel
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, Cagliari, Italy.
| | - Lucia Frau
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy.
| | - Paul B Huleatt
- Institute of Chemical Engineering and Science, A*STAR, 8 Biomedical Grove, Neuros, Singapore, 138665, Singapore.
| | - Mui-Ling Khoo
- Institute of Chemical Engineering and Science, A*STAR, 8 Biomedical Grove, Neuros, Singapore, 138665, Singapore.
| | - Christina L L Chai
- Institute of Chemical Engineering and Science, A*STAR, 8 Biomedical Grove, Neuros, Singapore, 138665, Singapore. .,Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore.
| | - Petra Dunkel
- Institute of Organic Chemistry, Semmelweis University, Budapest, Hungary.
| | - Peter Mátyus
- Institute of Organic Chemistry, Semmelweis University, Budapest, Hungary.
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy. .,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy.
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
30
|
Heng Y, Zhang QS, Mu Z, Hu JF, Yuan YH, Chen NH. Ginsenoside Rg1 attenuates motor impairment and neuroinflammation in the MPTP-probenecid-induced parkinsonism mouse model by targeting α-synuclein abnormalities in the substantia nigra. Toxicol Lett 2015; 243:7-21. [PMID: 26723869 DOI: 10.1016/j.toxlet.2015.12.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/14/2015] [Accepted: 12/21/2015] [Indexed: 11/27/2022]
Abstract
Parkinson's disease (PD) is pathologically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of aggregated α-synuclein in specific central nervous system (CNS) regions. Disease development is attributed to α-synuclein abnormalities, particularly aggregation and phosphorylation. The ginsenoside Rg1, an active component of ginseng, possesses neuroprotective and anti-inflammatory effects. The purpose of the present study was to evaluate these activities of Rg1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mouse model for the first time and to elucidate the underlying mechanisms. Oral treatment with Rg1 significantly attenuated the high MPTP-induced mortality, behavior defects, loss of dopamine neurons and abnormal ultrastructure changes in the SNpc. Other assays indicated that the protective effect of Rg1 may be mediated by its anti-neuroinflammatory properties. Rg1 regulated MPTP-induced reactive astrocytes and microglia and decreased the release of cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the SNpc. Rg1 also alleviated the unusual MPTP-induced increase in oligomeric, phosphorylated and disease-related α-synuclein in the SNpc. In conclusion, Rg1 protects dopaminergic neurons, most likely by reducing aberrant α-synuclein-mediated neuroinflammation, and holds promise for PD therapeutics.
Collapse
Affiliation(s)
- Yang Heng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qiu-Shuang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zheng Mu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jin-Feng Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
31
|
Muñoz-Manchado AB, Villadiego J, Romo-Madero S, Suárez-Luna N, Bermejo-Navas A, Rodríguez-Gómez JA, Garrido-Gil P, Labandeira-García JL, Echevarría M, López-Barneo J, Toledo-Aral JJ. Chronic and progressive Parkinson's disease MPTP model in adult and aged mice. J Neurochem 2015; 136:373-87. [DOI: 10.1111/jnc.13409] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/14/2015] [Accepted: 10/11/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Ana B. Muñoz-Manchado
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Javier Villadiego
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Sonia Romo-Madero
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Nela Suárez-Luna
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Alfonso Bermejo-Navas
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - José A. Rodríguez-Gómez
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Pablo Garrido-Gil
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Ciencias Morfológicas; Universidad de Santiago de Compostela; CIMUS; Instituto de Investigación Sanitaria (IDIS); Santiago de Compostela Spain
| | - José L. Labandeira-García
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Ciencias Morfológicas; Universidad de Santiago de Compostela; CIMUS; Instituto de Investigación Sanitaria (IDIS); Santiago de Compostela Spain
| | - Miriam Echevarría
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| | - Juan J. Toledo-Aral
- Instituto de Biomedicina de Sevilla-IBiS; HUVR/Universidad de Sevilla/CSIC; Sevilla Spain
- Departamento de Fisiología Médica y Biofísica; Universidad de Sevilla; Sevilla Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
| |
Collapse
|
32
|
Van Kampen JM, Baranowski DC, Robertson HA, Shaw CA, Kay DG. The Progressive BSSG Rat Model of Parkinson's: Recapitulating Multiple Key Features of the Human Disease. PLoS One 2015; 10:e0139694. [PMID: 26439489 PMCID: PMC4595214 DOI: 10.1371/journal.pone.0139694] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/16/2015] [Indexed: 12/21/2022] Open
Abstract
The development of effective neuroprotective therapies for Parkinson's disease (PD) has been severely hindered by the notable lack of an appropriate animal model for preclinical screening. Indeed, most models currently available are either acute in nature or fail to recapitulate all characteristic features of the disease. Here, we present a novel progressive model of PD, with behavioural and cellular features that closely approximate those observed in patients. Chronic exposure to dietary phytosterol glucosides has been found to be neurotoxic. When fed to rats, β-sitosterol β-d-glucoside (BSSG) triggers the progressive development of parkinsonism, with clinical signs and histopathology beginning to appear following cessation of exposure to the neurotoxic insult and continuing to develop over several months. Here, we characterize the progressive nature of this model, its non-motor features, the anatomical spread of synucleinopathy, and response to levodopa administration. In Sprague Dawley rats, chronic BSSG feeding for 4 months triggered the progressive development of a parkinsonian phenotype and pathological events that evolved slowly over time, with neuronal loss beginning only after toxin exposure was terminated. At approximately 3 months following initiation of BSSG exposure, animals displayed the early emergence of an olfactory deficit, in the absence of significant dopaminergic nigral cell loss or locomotor deficits. Locomotor deficits developed gradually over time, initially appearing as locomotor asymmetry and developing into akinesia/bradykinesia, which was reversed by levodopa treatment. Late-stage cognitive impairment was observed in the form of spatial working memory deficits, as assessed by the radial arm maze. In addition to the progressive loss of TH+ cells in the substantia nigra, the appearance of proteinase K-resistant intracellular α-synuclein aggregates was also observed to develop progressively, appearing first in the olfactory bulb, then the striatum, the substantia nigra and, finally, hippocampal and cortical regions. The slowly progressive nature of this model, together with its construct, face and predictive validity, make it ideal for the screening of potential neuroprotective therapies for the treatment of PD.
Collapse
Affiliation(s)
- Jackalina M. Van Kampen
- Neurodyn Inc., 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
- Dept. Biomedical Science, University of Prince Edward Island, 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
- * E-mail:
| | - David C. Baranowski
- Neurodyn Inc., 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
| | - Harold A. Robertson
- Neurodyn Inc., 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
- Dept. Biomedical Science, University of Prince Edward Island, 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
| | - Christopher A. Shaw
- Dept. Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Denis G. Kay
- Neurodyn Inc., 550 University Ave, Charlottetown, Prince Edward Island, C1A 4P3, Canada
| |
Collapse
|
33
|
Chronic MPTP treatment produces hyperactivity in male mice which is not alleviated by concurrent trehalose treatment. Behav Brain Res 2015; 292:68-78. [DOI: 10.1016/j.bbr.2015.05.057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/23/2015] [Indexed: 11/19/2022]
|
34
|
Maiti P, Gregg LC, McDonald MP. MPTP-induced executive dysfunction is associated with altered prefrontal serotonergic function. Behav Brain Res 2015; 298:192-201. [PMID: 26393431 DOI: 10.1016/j.bbr.2015.09.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/14/2015] [Accepted: 09/09/2015] [Indexed: 02/09/2023]
Abstract
In Parkinson's disease, cognitive deficits manifest as fronto-striatally-mediated executive dysfunction, with impaired attention, planning, judgment, and impulse control. We examined changes in executive function in mice lesioned with subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) using a 3-choice serial reaction-time (SRT) task, which included measures of sustained attention and impulse control. Each trial of the baseline SRT task comprised a pseudo-random pre-cue period ranging from 3 to 8 s, followed by a 1-s cue duration. MPTP impaired all measures of impulsive behavior acutely, but with additional training their performance normalized to saline control levels. When challenged with shorter cue durations, MPTP-lesioned mice had significantly slower reaction times than wild-type mice. When challenged with longer pre-cue times, the MPTP-lesioned mice exhibited a loss of impulse control at the longer durations. In lesioned mice, striatal dopamine was depleted by 54% and the number of tyrosine-hydroxylase-positive neurons in the substantia nigra pars compacta was reduced by 75%. Serotonin (5-HT) was unchanged in the striatum and prefrontal cortex (PFC), but the ratio of 5-hydroxyindolacetic acid (5-HIAA) to 5-HT was significantly reduced in the MPTP group in the PFC. In lesioned mice, prefrontal 5-HIAA/5-HT was significantly correlated with the executive impairments and striatal norepinephrine was associated with slower reaction times. None of the neurochemical measures was significantly associated with behavior in saline-treated controls. Taken together, these results show that prefrontal 5-HT turnover may play a pivotal role in MPTP-induced executive dysfunction.
Collapse
Affiliation(s)
- Panchanan Maiti
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Laura C Gregg
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Michael P McDonald
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
35
|
Khairnar A, Latta P, Drazanova E, Ruda-Kucerova J, Szabó N, Arab A, Hutter-Paier B, Havas D, Windisch M, Sulcova A, Starcuk Z, Rektorova I. Diffusion Kurtosis Imaging Detects Microstructural Alterations in Brain of α-Synuclein Overexpressing Transgenic Mouse Model of Parkinson’s Disease: A Pilot Study. Neurotox Res 2015; 28:281-9. [DOI: 10.1007/s12640-015-9537-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 06/29/2015] [Indexed: 12/12/2022]
|
36
|
Dietary Supplementation of Walnut Partially Reverses 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine Induced Neurodegeneration in a Mouse Model of Parkinson’s Disease. Neurochem Res 2015; 40:1283-93. [DOI: 10.1007/s11064-015-1593-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/21/2015] [Accepted: 04/28/2015] [Indexed: 12/21/2022]
|
37
|
Lecca D, Nevin DK, Mulas G, Casu MA, Diana A, Rossi D, Sacchetti G, Fayne D, Carta AR. Neuroprotective and anti-inflammatory properties of a novel non-thiazolidinedione PPARγ agonist in vitro and in MPTP-treated mice. Neuroscience 2015; 302:23-35. [PMID: 25907448 DOI: 10.1016/j.neuroscience.2015.04.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/12/2015] [Accepted: 04/11/2015] [Indexed: 11/29/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)γ is a potential pharmacological target for disease-modification in Parkinson's disease (PD), mainly acting by modulating the neuroinflammatory response. However, currently available agonists thiazolidinediones (TZDs) present limitations due to safety concerns. We evaluated a novel thiobarbituric-like compound MDG548, which acts as a functional PPARγ agonist displaying higher and selective binding affinity as compared to TZDs. Neuroprotection by MDG548 was tested in vitro and in a mouse MPTP model of PD, and neuroinflammation was investigated as a putative underlying mechanism. Viability assay on rat cortical neurons showed lack of cytotoxic effect in the dose-range of 100 nM-10 μM, which was therefore used for testing in vitro protection against H2O2 and MPP+ neurotoxicity. MDG548 dose-dependently increased cell viability of rat cortical neurons co-treated with H2O2 or pre-exposed to MDG548 prior to H2O2. Moreover, MDG548 induced neuroprotection in MPP+-treated PC12 cells. NF-kB activation was investigated to assess anti-inflammatory activity. MDG548 dose-dependently decreased NF-kB activation induced by LPS (100 ng/100ml) in HEK-Blue-hTLR4 cells. Given the supposed cancer risk of other PPARγ agonists, Ames test for genotoxicity was performed in Salmonella typhimurium TA100 and TA98 strains, showing that MDG548 was not genotoxic. In vivo, BL/6J mice were treated with MPTP (20mg/kg i.p. once/day for 4 days) in association with saline or MDG548 (2, 5, 10 mg/kg i.p.). Stereological counting showed that MDG548 prevented the MPTP-induced reduction in TH-positive cells in the substantia nigra compacta (SNc) at all doses tested. Moreover, MDG548 reduced reactive microglia and iNOS induction in the SNc. MDG548, being a non-TZD compound with high PPARγ affinity, void of genotoxicity, and with in vitro as well as in vivo neuroprotective properties, provides a promising alternative in the search for safer PPARγ agonists to be tested as potential disease-modifying drugs in PD.
Collapse
Affiliation(s)
- D Lecca
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - D K Nevin
- School of Biochemistry & Immunology, Trinity College, Dublin, Ireland
| | - G Mulas
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - M A Casu
- CNR-Institute of Translational Pharmacology, U.O.S. of Cagliari, Italy
| | - A Diana
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - D Rossi
- Department of Life Science and Biotechnology, University of Ferrara, Italy
| | - G Sacchetti
- Department of Life Science and Biotechnology, University of Ferrara, Italy
| | | | - A R Carta
- Department of Biomedical Sciences, University of Cagliari, Italy
| |
Collapse
|
38
|
Bao XQ, Wu LY, Wang XL, Sun H, Zhang D. Squamosamide derivative FLZ protected tyrosine hydroxylase function in a chronic MPTP/probenecid mouse model of Parkinson's disease. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:549-56. [PMID: 25678053 DOI: 10.1007/s00210-015-1094-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/19/2015] [Indexed: 05/28/2023]
Abstract
Parkinson's disease (PD) is a chronic, progressive neurodegenerative disorder characterized by motor impairments and loss of dopaminergic neurons in the substantia nigra. FLZ (formulated as: N-2-(4-hydroxy-phenyl)-ethyl]-2-(2, 5-dimethoxy-phenyl)-3-(3-methoxy-4-hydroxy-phenyl)-acrylamide) is a novel synthetic derivative of squamosamide from a Chinese herb and has been proven to protect dopaminergic neurons in subacute PD models. However, whether FLZ has a neuroprotective effect on chronic PD model is still unknown. The present study was designed to verify the neuroprotection of FLZ on chronic PD mouse model induced by MPTP combined with probenecid (MPTP/p). The results showed that treatment of mice with FLZ for 9 weeks significantly improved motor behavior and dopaminergic neuronal function of mice injected with MPTP/p. The beneficial effects of FLZ attributed to the elevation of dopaminergic neuron number, dopamine level, and tyrosine hydroxylase (TH) activity, as well as decrease of α-synuclein (α-Syn) expression, α-Syn phosphorylation, nitration, and aggregation. Moreover, FLZ decreased the interaction between α-Syn and TH, which eventually improved dopaminergic neuronal function. Mechanistic study demonstrated that FLZ increased Akt and mTOR phosphorylation, suggesting that FLZ activated Akt/mTOR signaling pathway and this might be involved in the neuroprotection of FLZ. The present results provided more elaborate in vivo evidences to support the neuroprotective effect of FLZ on dopaminergic neurons of chronic PD mouse model and the potential of FLZ to be developed as new drug to treat PD.
Collapse
Affiliation(s)
- Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,
| | | | | | | | | |
Collapse
|
39
|
McDonald MP. Methods and Models of the Nonmotor Symptoms of Parkinson Disease. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
40
|
Cote SR, Kuzhikandathil EV. Chronic levodopa treatment alters expression and function of dopamine D3 receptor in the MPTP/p mouse model of Parkinson's disease. Neurosci Lett 2014; 585:33-7. [PMID: 25445374 DOI: 10.1016/j.neulet.2014.11.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/11/2014] [Accepted: 11/15/2014] [Indexed: 01/25/2023]
Abstract
Chronic treatment with levodopa or antipsychotics results in manifestation of side-effects such as dyskinesia which correlates with changes in expression and function of receptors and signaling proteins. Previous studies have suggested a role for the dopamine D3 receptor in Parkinson's disease (PD) and tardive dyskinesia. Yet the expression and signaling function of D3 receptor in these disorders is not well understood. Here we tested the hypothesis that chronic levodopa treatment alters both expression and function of D3 receptors in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine plus probenecid (MPTP/p) mouse model of PD. drd3-EGFP reporter mice were injected biweekly with saline or MPTP and probenecid for a 5-week period. During the last two weeks of the 5-week period, the mice were administered saline or levodopa twice daily. Locomotor activity was measured during the treatment period. D3 receptor expression was determined by western blot analysis. D3 receptor signaling function was determined at tissue and single cell level by measuring the activation of D3 receptor-mitogen activated protein kinase (MAPK) pathway. The drd3-EGFP mice administered MPTP/p exhibited akinesia/bradykinesia. Expression of D3 receptor protein in the dorsal striatum specifically increased in the MPTP/p-treated mice administered levodopa. In the dorsal striatum of levodopa and MPTP/p-treated drd3-EGFP mice, administration of a D3 receptor-selective dose of agonist, PD128907, failed to activate D3 receptor-MAPK signaling. These results suggest that MPTP-induced lesion and chronic levodopa treatment alters D3 receptor expression and function in the dorsal striatum which could contribute to the development of dyskinesias and other motor side-effects.
Collapse
Affiliation(s)
- Samantha R Cote
- Department of Pharmacology and Physiology, Rutgers-New Jersey Medical School, 185 South Orange Avenue, MSB, I-647, Newark, NJ 07103, USA
| | - Eldo V Kuzhikandathil
- Department of Pharmacology and Physiology, Rutgers-New Jersey Medical School, 185 South Orange Avenue, MSB, I-647, Newark, NJ 07103, USA.
| |
Collapse
|
41
|
Pisanu A, Lecca D, Mulas G, Wardas J, Simbula G, Spiga S, Carta AR. Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson's disease. Neurobiol Dis 2014; 71:280-91. [DOI: 10.1016/j.nbd.2014.08.011] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 11/25/2022] Open
|
42
|
Abstract
Units of dendritic branches called dendritic spines represent more than simply decorative appendages of the neuron and actively participate in integrative functions of “spinous” nerve cells thereby contributing to the general phenomenon of synaptic plasticity. In animal models of drug addiction, spines are profoundly affected by treatments with drugs of abuse and represent important sub cellular markers which interfere deeply into the physiology of the neuron thereby providing an example of the burgeoning and rapidly increasing interest in “structural plasticity”. Medium Spiny Neurons (MSNs) of the Nucleus Accumbens (Nacc) show a reduced number of dendritic spines and a decrease in TH-positive terminals upon withdrawal from opiates, cannabinoids and alcohol. The reduction is localized “strictly” to second order dendritic branches where dopamine (DA)-containing terminals, impinging upon spines, make synaptic contacts. In addition, long-thin spines seems preferentially affected raising the possibility that cellular learning of these neurons may be selectively hampered. These findings suggest that dendritic spines are affected by drugs widely abused by humans and provide yet another example of drug-induced aberrant neural plasticity with marked reflections on the physiology of synapses, system structural organization, and neuronal circuitry remodeling.
Collapse
Affiliation(s)
- Saturnino Spiga
- Department of Animal Biology and Ecology, University of Cagliari Cagliari, Italy
| | - Giovanna Mulas
- Department of Animal Biology and Ecology, University of Cagliari Cagliari, Italy ; "G.Minardi" Laboratory of Cognitive Neuroscience, Department of Chemistry and Pharmacy, University of Sassari Sassari, Italy
| | - Francesca Piras
- Department of Animal Biology and Ecology, University of Cagliari Cagliari, Italy ; Department of Natural Science and the Territory, University of Sassari Sassari, Italy
| | - Marco Diana
- "G.Minardi" Laboratory of Cognitive Neuroscience, Department of Chemistry and Pharmacy, University of Sassari Sassari, Italy
| |
Collapse
|
43
|
Selvakumar GP, Janakiraman U, Essa MM, Justin Thenmozhi A, Manivasagam T. Escin attenuates behavioral impairments, oxidative stress and inflammation in a chronic MPTP/probenecid mouse model of Parkinson's disease. Brain Res 2014; 1585:23-36. [PMID: 24657313 DOI: 10.1016/j.brainres.2014.03.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/24/2014] [Accepted: 03/07/2014] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that results mainly due to the death of dopaminergic neurons in the substantia nigra (SN), and subsequently has an effect on one's motor function and coordination. The current investigation explored the neuroprotective potential of escin, a natural triterpene-saponin on chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p) induced mouse model of PD. Administration of MPTP led to the depleted striatal dopamine content, impaired patterns of behavior, enhanced oxidative stress and diminished expression of tyrosine hydroxylase (TH), dopamine transporter (DAT) and vesicular monoamine transporter-2 (VMAT-2). The expressions of interleukin-6 and -10, glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor protein-1 (IBA-1), tumor necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) in SN were also enhanced. Oral treatment of escin significantly attenuated MPTP/p induced dopaminergic markers depletion, physiological abnormalities, oxidative stress and inhibit neuroinflammatory cytokine expressions in SN. The result of our study confirmed that escin mediated its protection against experimental PD through its antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
| | - Udaiyappan Janakiraman
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamilnadu, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | | | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamilnadu, India.
| |
Collapse
|
44
|
Kim M, Cho KH, Shin MS, Lee JM, Cho HS, Kim CJ, Shin DH, Yang HJ. Berberine prevents nigrostriatal dopaminergic neuronal loss and suppresses hippocampal apoptosis in mice with Parkinson's disease. Int J Mol Med 2014; 33:870-8. [PMID: 24535622 DOI: 10.3892/ijmm.2014.1656] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/10/2013] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the selective loss of nigral dopaminergic neurons and a reduction in striatal dopaminergic fibers, which result in tremors, rigidity, bradykinesia and gait disturbance. In addition to motor dysfunction, dementia is a widely recognized symptom of patients with PD. Berberine, an isoquinoline alkaloid isolated from Berberis vulgaris L., is known to exert anxiolytic, analgesic, anti-inflammatory, antipsychotic, antidepressant and anti-amnesic effects. In the present study, we investigated the effects of berberine on short-term memory in relation to dopamine depletion and hippocampal neurogenesis using a mouse model of PD, induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/P) treatment. Mice in the berberine-treated groups were orally administered berberine once a day for a total of 5 weeks. Our results revealed that the injection of MPTP/P induced dopaminergic neuronal death in the substantia nigra and fiber loss in the striatum. This resulted in impaired motor balance and coordination, as assessed by the beam walking test. We further demonstrated that MPTP/P-induced apoptosis in the hippocampus deteriorated short-term memory, as shown by the step-down avoidance task. By contrast, neurogenesis in the hippocampal dentate gyrus, which is a compensatory adaptive response to excessive apoptosis, was increased upon PD induction. However, treatment with berberine enhanced motor balance and coordination by preventing dopaminergic neuronal damage. Treatment with berberine also improved short-term memory by inhibiting apoptosis in the hippocampus. Berberine demonstrated maximal potency at 50 mg/kg. Based on these data, treatment with berberine may serve as a potential therapeutic strategy for the alleviation of memory impairment and motor dysfunction in patients with PD.
Collapse
Affiliation(s)
- Mia Kim
- Department of Cardiovascular and Neurologic Diseases (Stroke Center), College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ki-Ho Cho
- Department of Cardiovascular and Neurologic Diseases (Stroke Center), College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Mal-Soon Shin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Jae-Min Lee
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Han-Sam Cho
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Dong-Hoon Shin
- Department of Food and Biotechnology, Graduate School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | - Hyeon Jeong Yang
- Department of Anesthesiology and Pain Medicine, CHA Bundang Medical Center, CHA University, Seongnam 463-721, Republic of Korea
| |
Collapse
|
45
|
Uncoupling protein 2 deficiency aggravates astrocytic endoplasmic reticulum stress and nod-like receptor protein 3 inflammasome activation. Neurobiol Aging 2014; 35:421-30. [DOI: 10.1016/j.neurobiolaging.2013.08.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 08/02/2013] [Accepted: 08/16/2013] [Indexed: 01/07/2023]
|
46
|
Wojtowicz AK, Szychowski KA, Kajta M. PPAR-γ agonist GW1929 but not antagonist GW9662 reduces TBBPA-induced neurotoxicity in primary neocortical cells. Neurotox Res 2013; 25:311-22. [PMID: 24132472 PMCID: PMC3936120 DOI: 10.1007/s12640-013-9434-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/15/2013] [Accepted: 10/02/2013] [Indexed: 11/30/2022]
Abstract
Tetrabromobisphenol A (2,2-bis(4-hydroxy-3,5-dibromophenyl)propane; TBBPA) is a widely used brominated flame retardant. TBBPA induces neuronal damage, but the mechanism by which this occurs is largely unknown. We studied the possible involvement of peroxisome proliferator-activated receptor gamma (PPAR-γ) in TBBPA-induced apoptosis and toxicity in mouse primary neuronal cell cultures. TBBPA enhanced both, caspase-3 activity and lactate dehydrogenase (LDH) release in neocortical cells after 6 and 24 h of exposition. These data were supported at the cellular level with Hoechst 33342 staining. Immunoblot analyses showed that, compared with control cells, 10 μM TBBPA decreased the expression of PPAR-γ protein in neocortical neurons after 1-24 h of exposure. Co-treatment with TBBPA and GW1929 inhibited the TBBPA-induced caspase-3 activity, apoptotic body formation, and LDH release as well as TBBPA-induced decrease in PPAR-γ protein expression. Thus, our data support neuroprotective potential of PPAR-γ agonists. The PPAR-γ antagonist GW9662 prevented the TBBPA-induced decrease in PPAR-γ protein level, but it potentiated TBBPA-induced apoptotic and neurotoxic effects, which suggest that the mechanism of TBBPA action in neuronal cells is not only PPAR-γ-dependent. Therefore, further studies of the mechanism of TBBPA action in the nervous system are needed.
Collapse
Affiliation(s)
- Anna K Wojtowicz
- Laboratory of Genomics and Biotechnology, Animal Sciences Faculty, University of Agriculture, Redzina 1B, 30-248, Krakow, Poland,
| | | | | |
Collapse
|
47
|
Taylor JM, Main BS, Crack PJ. Neuroinflammation and oxidative stress: Co-conspirators in the pathology of Parkinson’s disease. Neurochem Int 2013; 62:803-19. [DOI: 10.1016/j.neuint.2012.12.016] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/20/2012] [Accepted: 12/26/2012] [Indexed: 12/21/2022]
|
48
|
Nasal inoculation with α-synuclein aggregates evokes rigidity, locomotor deficits and immunity to such misfolded species as well as dopamine. Behav Brain Res 2013; 243:205-12. [DOI: 10.1016/j.bbr.2013.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/06/2013] [Accepted: 01/10/2013] [Indexed: 12/26/2022]
|
49
|
Abstract
Parkinson's disease (PD) is characterized by a progressive degeneration of dopamine (DA) neurons and a chronic loss of motor functions. The investigation of progressive degenerative mechanisms and possible neuroprotective approaches for PD depends upon the development of an experimental animal model that reproduces the neuropathology observed in humans. This chapter describes the generation of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTPp) chronic mouse model of PD. This model displays key features of PD, including impairment of motor and olfactory functions associated with partial loss of tyrosine hydroxylase-positive neurons and DA levels in the brain. The MPTPp mouse model provides an important tool for the study of mechanisms contributing to the pathological dysfunction of PD at the cellular and whole animal level.
Collapse
Affiliation(s)
- Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | | | | |
Collapse
|
50
|
Liu Y, Pan W, Yang S, Wu X, Wu J, Ma J, Yuan Z, Meng S. Interleukin-22 protects rat PC12 pheochromocytoma cells from serum deprivation-induced cell death. Mol Cell Biochem 2012; 371:137-46. [PMID: 22983826 DOI: 10.1007/s11010-012-1430-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 08/03/2012] [Indexed: 12/13/2022]
Abstract
Interleukin-22 (IL-22), an IL-10 family cytokine, mediates the crosstalk between leukocytes and epithelial cells. Previous studies reported that IL-22 expresses in mouse brain, and the rat PC12 cells are responsive to IL-22 stimulation. However, the biological roles of IL-22 in neuronal cells remain largely unknown. We show here that IL-22 activates Stat3, p38 mitogen-activated protein kinases (MAPK), and Akt pathways and inhibits Erk/MAPK pathway in naïve PC12 cells. We further demonstrate that IL-22 protects naïve PC12 cells from serum starvation-induced cell death via the Jak1/Stat3 and Akt pathways. We also show that IL-22 has no effects on naïve PC12 cell proliferation and cannot protect naïve PC12 cells from 1-methyl-4-phenylpyridinium (MPP(+))-induced cytotoxicity. However, IL-22 exerts a dose-dependent protective effect on MPP(+)-induced neurodegeneration in nerve growth factor-differentiated PC12 cells. Overall, our data suggest that IL-22 might play a role in neurological processes. To our knowledge, this is the first report showing that IL-22 confers a neuroprotective function, which may provide a new therapeutic option for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yongchun Liu
- Northern Jiangsu People's Hospital, Nantong West Road No. 98, Yangzhou 225001, China
| | | | | | | | | | | | | | | |
Collapse
|