1
|
González-Cota AL, Martínez-Flores D, Rosendo-Pineda MJ, Vaca L. NMDA receptor-mediated Ca 2+ signaling: Impact on cell cycle regulation and the development of neurodegenerative diseases and cancer. Cell Calcium 2024; 119:102856. [PMID: 38408411 DOI: 10.1016/j.ceca.2024.102856] [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: 09/11/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
NMDA receptors are Ca2+-permeable ligand-gated ion channels that mediate fast excitatory transmission in the central nervous system. NMDA receptors regulate the proliferation and differentiation of neural progenitor cells and also play critical roles in neural plasticity, memory, and learning. In addition to their physiological role, NMDA receptors are also involved in glutamate-mediated excitotoxicity, which results from excessive glutamate stimulation, leading to Ca2+ overload, and ultimately to neuronal death. Thus, NMDA receptor-mediated excitotoxicity has been linked to several neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, dementia, and stroke. Interestingly, in addition to its effects on cell death, aberrant expression or activation of NMDA receptors is also involved in pathological cellular proliferation, and is implicated in the invasion and proliferation of various types of cancer. These disorders are thought to be related to the contribution of NMDA receptors to cell proliferation and cell death through cell cycle modulation. This review aims to discuss the evidence implicating NMDA receptor activity in cell cycle regulation and the link between aberrant NMDA receptor activity and the development of neurodegenerative diseases and cancer due to cell cycle dysregulation. The information presented here will provide insights into the signaling pathways and the contribution of NMDA receptors to these diseases, and suggests that NMDA receptors are promising targets for the prevention and treatment of these diseases, which are leading causes of death and disability worldwide.
Collapse
Affiliation(s)
- Ana L González-Cota
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Daniel Martínez-Flores
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Margarita Jacaranda Rosendo-Pineda
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Luis Vaca
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico.
| |
Collapse
|
2
|
Emmi A, Campagnolo M, Stocco E, Carecchio M, Macchi V, Antonini A, De Caro R, Porzionato A. Neurotransmitter and receptor systems in the subthalamic nucleus. Brain Struct Funct 2023; 228:1595-1617. [PMID: 37479801 PMCID: PMC10471682 DOI: 10.1007/s00429-023-02678-z] [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: 02/10/2023] [Accepted: 07/02/2023] [Indexed: 07/23/2023]
Abstract
The Subthalamic Nucleus (STh) is a lens-shaped subcortical structure located ventrally to the thalamus, that despite being embryologically derived from the diencephalon, is functionally implicated in the basal ganglia circuits. Because of this strict structural and functional relationship with the circuits of the basal ganglia, the STh is a current target for deep brain stimulation, a neurosurgical procedure employed to alleviate symptoms in movement disorders, such as Parkinson's disease and dystonia. However, despite the great relevance of this structure for both basal ganglia physiology and pathology, the neurochemical and molecular anatomy of the STh remains largely unknown. Few studies have specifically addressed the detection of neurotransmitter systems and their receptors within the structure, and even fewer have investigated their topographical distribution. Here, we have reviewed the scientific literature on neurotransmitters relevant in the STh function of rodents, non-human primates and humans including glutamate, GABA, dopamine, serotonin, noradrenaline with particular focus on their subcellular, cellular and topographical distribution. Inter-species differences were highlighted to provide a framework for further research priorities, particularly in humans.
Collapse
Affiliation(s)
- Aron Emmi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Marta Campagnolo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Elena Stocco
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Veronica Macchi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| | - Raffaele De Caro
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy.
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy.
| | - Andrea Porzionato
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padua, Italy
| |
Collapse
|
3
|
Bourque M, Morissette M, Conquet F, Charvin D, Di Paolo T. Foliglurax, a positive allosteric modulator of the metabotrophic glutamate receptor 4, protects dopaminergic neurons in MPTP-lesioned male mice. Brain Res 2023; 1809:148349. [PMID: 36972837 DOI: 10.1016/j.brainres.2023.148349] [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: 01/31/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/28/2023]
Abstract
Overactivity of the corticostriatal glutamatergic pathway is documented in Parkinson's disease (PD) and stimulation of presynaptic metabotropic glutamate (mGlu) receptors 4 on these striatal afferents inhibits glutamate release normalizing neuronal activity in the basal ganglia. Moreover, mGlu4 receptors are also expressed in glial cells and are able to modulate glial function making this receptor a potential target for neuroprotection. Hence, we investigated whether foliglurax, a positive allosteric modulator of mGlu4 receptors with high brain exposure after oral administration, has neuroprotective effects in MPTP mice to model early PD. Male mice were treated daily from day 1 to 10 with 1, 3 or 10 mg/kg of foliglurax and administered MPTP on the 5th day then euthanized on the 11th day. Dopamine neuron integrity was assessed with measures of striatal dopamine and its metabolites levels, striatal and nigral dopamine transporter (DAT) binding and inflammation with markers of striatal astrocytes (GFAP) and microglia (Iba1). MPTP lesion produced a decrease in dopamine, its metabolites and striatal DAT specific binding that was prevented by treatment with 3 mg/kg of foliglurax, whereas 1 and 10 mg/kg had no beneficial effect. MPTP mice had increased levels of GFAP; foliglurax treatment (3 mg/kg) prevented this increase. Iba1 levels were unchanged in MPTP mice compared to control mice. There was a negative correlation between dopamine content and GFAP levels. Our results show that positive allosteric modulation of mGlu4 receptors with foliglurax provided neuroprotective effects in the MPTP mouse model of PD.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | | | | | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
4
|
Wal P, Dwivedi J, Wal A, Vig H, Singh Y. Detailed insight into the pathophysiology and the behavioral complications associated with the Parkinson's disease and its medications. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The loss of dopamine neurons in the substantia nigra, as well as other mostly catecholaminergic neurons, causes many of the motor symptoms that define Parkinson's disease. Parkinson's disease is commonly thought of as a movement disorder, the significant prevalence of psychiatric complications such as cognitive impairment, and psychosis suggests it should be considered a neuropsychiatric illness, and all behavioral complications are linked to growing disability and the medication.
Main body
Apart from the disease-induced abnormalities, there are several other side effects of the disease and also from the medication used to prevent the disease. This article focuses on the pathogenesis of Parkinson’s disease and also the behavioral abnormalities caused by the disease and its medication. The study's data were gathered by searching several review articles and research papers from a variety of sources, including Elsevier, PubMed, Research Gate, Journal of Pharmaceutical Science, etc., from the year 1985 to 2021. Parkinson's disease is a neurodegenerative disease caused by a variety of complex processes. It is responsible not just for motor symptoms, but also for a variety of behavioral symptoms that can arise as a result of the disease and/or medication.
Conclusion
Only symptomatic drugs are available; thus, finding treatments that directly address the disease mechanisms causing Parkinson’s disease is essential. To alleviate the disease's burden on patients and their families, better treatments for the neuropsychiatric repercussions of Parkinson's disease are required.
Graphical Abstract
Collapse
|
5
|
Schrank S, Barrington N, Stutzmann GE. Calcium-Handling Defects and Neurodegenerative Disease. Cold Spring Harb Perspect Biol 2020; 12:a035212. [PMID: 31427373 PMCID: PMC7328457 DOI: 10.1101/cshperspect.a035212] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Calcium signaling is critical to neuronal function and regulates highly diverse processes such as gene transcription, energy production, protein handling, and synaptic structure and function. Because there are many common underlying calcium-mediated pathological features observed across several neurological conditions, it has been proposed that neurodegenerative diseases have an upstream underlying calcium basis in their pathogenesis. With certain diseases such as Alzheimer's, Parkinson's, and Huntington's, specific sources of calcium dysregulation originating from distinct neuronal compartments or channels have been shown to have defined roles in initiating or sustaining disease mechanisms. Herein, we will review the major hallmarks of these diseases, and how they relate to calcium dysregulation. We will then discuss neuronal calcium handling throughout the neuron, with special emphasis on channels involved in neurodegeneration.
Collapse
Affiliation(s)
- Sean Schrank
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Nikki Barrington
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois 60064
| |
Collapse
|
6
|
Szlufik S, Duszynska-Lysak K, Przybyszewski A, Laskowska-Levy I, Drzewinska A, Dutkiewicz J, Mandat T, Habela P, Koziorowski D. The potential neuromodulatory impact of subthalamic nucleus deep brain stimulation on Parkinson's disease progression. J Clin Neurosci 2020; 73:150-154. [PMID: 32001113 DOI: 10.1016/j.jocn.2019.12.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/30/2019] [Indexed: 11/21/2022]
Abstract
INTRODUCTION STN-DBS has been claimed to change progressionsymptomsin animal models of PD, but information is lacking about the possible neuromodulatory role of STN-DBS in humans. The aim of this prospective controlled study was to evaluate the long-term impact of STN-DBS on motor disabilities and cognitive impairment in PD patients in comparison to Best-Medical-Therapy (BMT) and Long-term-Post-Operative (POP) groups. MATERIAL AND METHODS Patients were divided into 3 groups: the BMT-group consisted of 20 patients treated only with pharmacotherapy, the DBS-group consisted of 20 PD patients who underwent bilateral STN-DBS (examined pre- and postoperatively) and the POP-group consisted of 14 long-term postoperative patients in median 30 month-time after DBS. UPDRS III scale was measured during 3 visits in 9 ± 2 months periods (V1, V2, V3) in total-OFF phase. Cognitive assessment was performed during each visit in total-ON phase. RESULTS The comparable UPDRS III OFF gain was observed in both BMT-group and POP-group evaluations (p < 0.05). UPDRS III OFF results in DBS-group revealed significant UPDRS III OFF increase in ΔV2-V1 assessment (p < 0.05) with no significant UPDRS III OFF alteration in ΔV3-V2 DBS-group evaluation (p > 0.05). Cognitive assessment revealed significant alterations between DBS-group and BMT-group in working memory, executive functions and learning abilities (p < 0.05). CONCLUSIONS The impact of STN-DBS on UPDRS III OFF score and cognitive alterations suggest its neuromodulatory role, mainly during the first 9-18 months after surgery.
Collapse
Affiliation(s)
- Stanislaw Szlufik
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Poland.
| | | | - Andrzej Przybyszewski
- Faculty of Information Technology, Polish Japanese Academy of Information Technology, Poland
| | - Ilona Laskowska-Levy
- Music Performance and Brain Laboratory, Department of Cognitive Psychology, University of Economics and Human Sciences, Warsaw, Poland; Psychotherapy Center, XIIIth Daily Department of Neurotic Disorders, Nowowiejski Hospital, Warsaw, Poland
| | - Agnieszka Drzewinska
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Poland
| | - Justyna Dutkiewicz
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Poland
| | - Tomasz Mandat
- Department of Neurosurgery, Maria Sklodowska Curie Memorial Oncology Center, Warsaw, Poland
| | - Piotr Habela
- Faculty of Information Technology, Polish Japanese Academy of Information Technology, Poland
| | - Dariusz Koziorowski
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Poland
| |
Collapse
|
7
|
Stayte S, Laloli KJ, Rentsch P, Lowth A, Li KM, Pickford R, Vissel B. The kainate receptor antagonist UBP310 but not single deletion of GluK1, GluK2, or GluK3 subunits, inhibits MPTP-induced degeneration in the mouse midbrain. Exp Neurol 2020; 323:113062. [PMID: 31513786 DOI: 10.1016/j.expneurol.2019.113062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/20/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022]
Abstract
The excitatory neurotransmitter glutamate is essential in basal ganglia motor circuits and has long been thought to contribute to cell death and degeneration in Parkinson's disease (PD). While previous research has shown a significant role of NMDA and AMPA receptors in both excitotoxicity and PD, the third class of ionotropic glutamate receptors, kainate receptors, have been less well studied. Given the expression of kainate receptor subunits GluK1-GluK3 in key PD-related brain regions, it has been suggested that GluK1-GluK3 may contribute to excitotoxic cell loss. Therefore the neuroprotective potential of the kainate receptor antagonist UBP310 in animal models of PD was investigated in this study. Stereological quantification revealed administration of UBP310 significantly increased survival of dopaminergic and total neuron populations in the substantia nigra pars compacta in the acute MPTP mouse model of PD. In contrast, UBP310 was unable to rescue MPTP-induced loss of dopamine levels or dopamine transporter expression in the striatum. Furthermore, deletion of GluK1, GluK2 or GluK3 had no effect on MPTP or UBP310-mediated effects across all measures. Interestingly, UBP310 did not attenuate cell loss in the midbrain induced by intrastriatal 6-OHDA toxicity. These results indicate UBP310 provides neuroprotection in the midbrain against MPTP neurotoxicity that is not dependent on specific kainate receptor subunits.
Collapse
Affiliation(s)
- Sandy Stayte
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, Australia; St Vincent's Centre for Applied Medical Research (AMR), Sydney, Darlinghurst, Australia; Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Kathryn J Laloli
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, Australia; St Vincent's Centre for Applied Medical Research (AMR), Sydney, Darlinghurst, Australia
| | - Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, Australia; St Vincent's Centre for Applied Medical Research (AMR), Sydney, Darlinghurst, Australia; Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Aimee Lowth
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Kong M Li
- Pharmacology Department, Bosch Institute, Sydney Medical School, The University of Sydney, Camperdown, Australia
| | - Russell Pickford
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Kensington, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, Australia; St Vincent's Centre for Applied Medical Research (AMR), Sydney, Darlinghurst, Australia; Garvan Institute of Medical Research, Darlinghurst, Australia.
| |
Collapse
|
8
|
Bhattacharya S, Ma Y, Dunn AR, Bradner JM, Scimemi A, Miller GW, Traynelis SF, Wichmann T. NMDA receptor blockade ameliorates abnormalities of spike firing of subthalamic nucleus neurons in a parkinsonian nonhuman primate. J Neurosci Res 2018; 96:1324-1335. [PMID: 29577359 PMCID: PMC5980712 DOI: 10.1002/jnr.24230] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/25/2018] [Accepted: 02/08/2018] [Indexed: 12/21/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ion channels comprising tetrameric assemblies of GluN1 and GluN2 receptor subunits that mediate excitatory neurotransmission in the central nervous system. Of the four different GluN2 subunits, the GluN2D subunit-containing NMDARs have been suggested as a target for antiparkinsonian therapy because of their expression pattern in some of the basal ganglia nuclei that show abnormal firing patterns in the parkinsonian state, specifically the subthalamic nucleus (STN). In this study, we demonstrate that blockade of NMDARs altered spike firing in the STN in a male nonhuman primate that had been rendered parkinsonian by treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. In accompanying experiments in male rodents, we found that GluN2D-NMDAR expression in the STN was reduced in acutely or chronically dopamine-depleted animals. Taken together, our data suggest that blockade of NMDARs in the STN may be a viable antiparkinsonian strategy, but that the ultimate success of this approach may be complicated by parkinsonism-associated changes in NMDAR expression in the STN.
Collapse
Affiliation(s)
| | - Yuxian Ma
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Amy R Dunn
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Joshua M Bradner
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Annalisa Scimemi
- Department of Biology, State University of New York at Albany, Albany, New York
| | - Gary W Miller
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
- Morris K. Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| |
Collapse
|
9
|
Lavaur J, Le Nogue D, Lemaire M, Pype J, Farjot G, Hirsch EC, Michel PP. The noble gas xenon provides protection and trophic stimulation to midbrain dopamine neurons. J Neurochem 2017; 142:14-28. [PMID: 28398653 PMCID: PMC5518208 DOI: 10.1111/jnc.14041] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 01/24/2023]
Abstract
Despite its low chemical reactivity, the noble gas xenon possesses a remarkable spectrum of biological effects. In particular, xenon is a strong neuroprotectant in preclinical models of hypoxic‐ischemic brain injury. In this study, we wished to determine whether xenon retained its neuroprotective potential in experimental settings that model the progressive loss of midbrain dopamine (DA) neurons in Parkinson's disease. Using rat midbrain cultures, we established that xenon was partially protective for DA neurons through either direct or indirect effects on these neurons. So, when DA neurons were exposed to l‐trans‐pyrrolidine‐2,4‐dicarboxylic acid so as to increase ambient glutamate levels and generate slow and sustained excitotoxicity, the effect of xenon on DA neurons was direct. The vitamin E analog Trolox also partially rescued DA neurons in this setting and enhanced neuroprotection by xenon. However, in the situation where DA cell death was spontaneous, the protection of DA neurons by xenon appeared indirect as it occurred through the repression of a mechanism mediated by proliferating glial cells, presumably astrocytes and their precursor cells. Xenon also exerted trophic effects for DA neurons in this paradigm. The effects of xenon were mimicked and improved by the N‐methyl‐d‐aspartate glutamate receptor antagonist memantine and xenon itself appeared to work by antagonizing N‐methyl‐d‐aspartate receptors. Note that another noble gas argon could not reproduce xenon effects. Overall, present data indicate that xenon can provide protection and trophic support to DA neurons that are vulnerable in Parkinson's disease. This suggests that xenon might have some therapeutic value for this disorder. ![]()
Collapse
Affiliation(s)
- Jérémie Lavaur
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Paris, France
| | - Déborah Le Nogue
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Paris, France
| | - Marc Lemaire
- Air Liquide Santé International, Medical R&D Paris, Saclay Research Center, Jouy-en Josas, France
| | - Jan Pype
- Air Liquide Santé International, Medical R&D Paris, Saclay Research Center, Jouy-en Josas, France
| | - Géraldine Farjot
- Air Liquide Santé International, Medical R&D Paris, Saclay Research Center, Jouy-en Josas, France
| | - Etienne C Hirsch
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Paris, France
| | - Patrick P Michel
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
10
|
Petrov D, Pedros I, de Lemos ML, Pallàs M, Canudas AM, Lazarowski A, Beas-Zarate C, Auladell C, Folch J, Camins A. Mavoglurant as a treatment for Parkinson's disease. Expert Opin Investig Drugs 2014; 23:1165-79. [PMID: 24960254 DOI: 10.1517/13543784.2014.931370] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION A major unresolved issue in the Parkinson's disease (PD) treatment is the development of l-DOPA-induced dyskinesias (LIDs) as a side effect of chronic L-DOPA administration. Currently, LIDs are managed in part by reducing the L-DOPA dose or by the administration of amantadine. However, this treatment is only partially effective. A potential strategy, currently under investigation, is the coadministration of metabotropic glutamate receptor 5 (mGluR5) negative allosteric modulators (NAMs) and L-DOPA; a treatment that results in the improvement of dyskinesia symptoms and that permits reductions in l-DOPA dosage frequency. AREAS COVERED The authors examine the role of mGluR5 in the pathophysiology of PD and the potential use of mGluR5 NAM as an adjuvant therapy together with a primary treatment with L-DOPA. Specifically, the authors look at the mavoglurant therapy and the evidence presented through preclinical and clinical trials. EXPERT OPINION Interaction between mGluR5 NAM and L-DOPA is an area of interest in PD research as concomitant treatment results in the improvement of LID symptoms in humans, thus enhancing the patient's quality of life. However, few months ago, Novartis decided to discontinue clinical trials of mavoglurant for the treatment of LID, due to the lack of efficacy demonstrated in trials NCT01385592 and NCT01491529, although no safety concerns were involved in this decision. Nevertheless, the potential application of mGluR5 antagonists as neuroprotective agents must be considered and further studies are warranted to better investigate their potential.
Collapse
Affiliation(s)
- Dmitry Petrov
- Universitat de Barcelona, Institut de Biomedicina (IBUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Unitat de Farmacologia I Farmacognòsia, Facultat de Farmàcia , Barcelona, Avda/Joan XXIII , Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Finlay C, Duty S. Therapeutic potential of targeting glutamate receptors in Parkinson's disease. J Neural Transm (Vienna) 2014; 121:861-80. [PMID: 24557498 DOI: 10.1007/s00702-014-1176-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/05/2014] [Indexed: 12/28/2022]
Abstract
Glutamate plays a complex role in many aspects of Parkinson's disease including the loss of dopaminergic neurons, the classical motor symptoms as well as associated non-motor symptoms and the treatment-related side effect, L-DOPA-induced dyskinesia. This widespread involvement opens up possibilities for glutamate-based therapies to provide a more rounded approach to treatment than is afforded by current dopamine replacement therapies. Beneficial effects of blocking postsynaptic glutamate transmission have already been noted in a range of preclinical studies using antagonists of NMDA receptors or negative allosteric modulators of metabotropic glutamate receptor 5 (mGlu5), while positive allosteric modulators of mGlu4 in particular, although at an earlier stage of investigation, also look promising. This review addresses each of the key features of Parkinson's disease in turn, summarising the contribution glutamate makes to that feature and presenting an up-to-date account of the potential for drugs acting at ionotropic or metabotropic glutamate receptors to provide relief. Whilst only a handful of these have progressed to clinical trials to date, notably NMDA and NR2B antagonists against motor symptoms and L-DOPA-induced dyskinesia, with mGlu5 negative allosteric modulators also against L-DOPA-induced dyskinesia, the mainly positive outcomes of these trials, coupled with supportive preclinical data for other strategies in animal models of Parkinson's disease and L-DOPA-induced dyskinesia, raise cautious optimism that a glutamate-based therapeutic approach will have significant impact on the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Clare Finlay
- Wolfson Centre for Age-Related Diseases, King's College London, WW1.28. Hodgkin Building, Guy's Campus, London, SE1 1UL, UK
| | | |
Collapse
|
12
|
Ambrosi G, Cerri S, Blandini F. A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. J Neural Transm (Vienna) 2014; 121:849-59. [DOI: 10.1007/s00702-013-1149-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
|
13
|
Targeting glutamate receptors to tackle the pathogenesis, clinical symptoms and levodopa-induced dyskinesia associated with Parkinson's disease. CNS Drugs 2012; 26:1017-32. [PMID: 23114872 DOI: 10.1007/s40263-012-0016-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The appearance of levodopa-induced dyskinesia (LID) and ongoing degeneration of nigrostriatal dopaminergic neurons are two key features of Parkinson's disease (PD) that current treatments fail to address. Increased glutamate transmission contributes to the motor symptoms in PD, to the striatal plasticity that underpins LID and to the progression of neurodegeneration through excitotoxic mechanisms. Glutamate receptors have therefore long been considered as potential targets for pharmacological intervention in PD, with emphasis on either blocking activation of 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid (AMPA), N-methyl-D-aspartate (NMDA) or excitatory metabotropic glutamate (mGlu) 5 receptors or promoting the activation of group II/III mGlu receptors. Following a brief summary of the role of glutamate in PD and LID, this article explores the current status of pharmacological studies in pre-clinical rodent and primate models through to clinical trials, where applicable, that support the potential of glutamate-based therapeutic interventions. To date, AMPA antagonists have shown good efficacy against LID in rat and primate models, but the failure of perampanel to lessen LID in clinical trials casts doubt on the translational potential of this approach. In contrast, antagonists selective for NR2B-containing NMDA receptors were effective against LID in animal models and in small-scale clinical trials, though observed adverse cognitive effects need addressing. So far, mGlu5 antagonists or negative allosteric modulators (NAMs) look set to become the first introduced for tackling LID, with AFQ-056 reported to exhibit good efficacy in phase II clinical trials. NR2B antagonists and mGlu5 NAMs may subsequently prove to also be effective disease-modifying agents if their protective effects in rat and primate models of PD, respectively, are replicated in the next stages of investigation. Finally, group III mGlu4 agonists or positive allosteric modulators (PAMs), although in the early pre-clinical stages of investigation, are showing good efficacy against motor symptoms, neurodegeneration and LID. It is anticipated that the recent development of mGlu4 PAMs with improved systemic bioavailability will facilitate progression of these agents into the primate model of PD where their potential can be further explored.
Collapse
|
14
|
Harnack D, Kupsch A. The impact of subthalamic deep brain stimulation on nigral neuroprotection-myth or reality? Neuromodulation 2012; 13:160-7. [PMID: 21992827 DOI: 10.1111/j.1525-1403.2010.00282.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE In the present review article we summarize available clinical and preclinical evidence, if modulation of the subthalamic nucleus (STN) could be a target for neuroprotection in Parkinson's disease (PD). BACKGROUND Chronic deep brain stimulation (DBS) of the STN has emerged as a powerful therapeutic alternative for the treatment of PD, ensuring stable symptom control for up to five years despite the progressive nature PD. MATERIALS AND METHODS Comparative review of literature in PuBMed available up to December 2008. RESULTS The assessment of neuroprotection has been proven difficult in the clinical situation, as medical or surgical therapeutic options that improve PD symptoms could be erroneously considered to be neuroprotective because of the difficulty of differentiating between symptomatic effects and potential neuromodulative disease-related effects of various treatment options applied in PD. The methodological limitations of clinical trials underline the importance of putative neuroprotective compounds to be tested in clinically driven preclinical studies. Thus, animal models, mimicking progressive nigrostriatal cell death, are indispensable to further advance the important issue of neuroprotection or neuromodulation following DBS. CONCLUSION Clear clinical evidence for STN-DBS-related neuroprotection in PD is missing. However, numerous preclinical studies show (and are discussed) that silencing of the STN via lesion or DBS may exert neuromodulative effects on nigral dopamine neurons.
Collapse
Affiliation(s)
- Daniel Harnack
- Department of Neurology, Charité, University Medicine Berlin, Berlin, Germany
| | | |
Collapse
|
15
|
Hsieh MH, Gu SL, Ho SC, Pawlak CR, Lin CL, Ho YJ, Lai TJ, Wu FY. Effects of MK-801 on recognition and neurodegeneration in an MPTP-induced Parkinson's rat model. Behav Brain Res 2012; 229:41-7. [DOI: 10.1016/j.bbr.2011.12.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 12/19/2011] [Accepted: 12/21/2011] [Indexed: 11/16/2022]
|
16
|
Ledonne A, Mango D, Bernardi G, Berretta N, Mercuri NB. A continuous high frequency stimulation of the subthalamic nucleus determines a suppression of excitatory synaptic transmission in nigral dopaminergic neurons recorded in vitro. Exp Neurol 2012; 233:292-302. [DOI: 10.1016/j.expneurol.2011.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/10/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
|
17
|
Hamza TH, Chen H, Hill-Burns EM, Rhodes SL, Montimurro J, Kay DM, Tenesa A, Kusel VI, Sheehan P, Eaaswarkhanth M, Yearout D, Samii A, Roberts JW, Agarwal P, Bordelon Y, Park Y, Wang L, Gao J, Vance JM, Kendler KS, Bacanu SA, Scott WK, Ritz B, Nutt J, Factor SA, Zabetian CP, Payami H. Genome-wide gene-environment study identifies glutamate receptor gene GRIN2A as a Parkinson's disease modifier gene via interaction with coffee. PLoS Genet 2011; 7:e1002237. [PMID: 21876681 PMCID: PMC3158052 DOI: 10.1371/journal.pgen.1002237] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 06/24/2011] [Indexed: 11/18/2022] Open
Abstract
Our aim was to identify genes that influence the inverse association of coffee with the risk of developing Parkinson's disease (PD). We used genome-wide genotype data and lifetime caffeinated-coffee-consumption data on 1,458 persons with PD and 931 without PD from the NeuroGenetics Research Consortium (NGRC), and we performed a genome-wide association and interaction study (GWAIS), testing each SNP's main-effect plus its interaction with coffee, adjusting for sex, age, and two principal components. We then stratified subjects as heavy or light coffee-drinkers and performed genome-wide association study (GWAS) in each group. We replicated the most significant SNP. Finally, we imputed the NGRC dataset, increasing genomic coverage to examine the region of interest in detail. The primary analyses (GWAIS, GWAS, Replication) were performed using genotyped data. In GWAIS, the most significant signal came from rs4998386 and the neighboring SNPs in GRIN2A. GRIN2A encodes an NMDA-glutamate-receptor subunit and regulates excitatory neurotransmission in the brain. Achieving P(2df) = 10(-6), GRIN2A surpassed all known PD susceptibility genes in significance in the GWAIS. In stratified GWAS, the GRIN2A signal was present in heavy coffee-drinkers (OR = 0.43; P = 6×10(-7)) but not in light coffee-drinkers. The a priori Replication hypothesis that "Among heavy coffee-drinkers, rs4998386_T carriers have lower PD risk than rs4998386_CC carriers" was confirmed: OR(Replication) = 0.59, P(Replication) = 10(-3); OR(Pooled) = 0.51, P(Pooled) = 7×10(-8). Compared to light coffee-drinkers with rs4998386_CC genotype, heavy coffee-drinkers with rs4998386_CC genotype had 18% lower risk (P = 3×10(-3)), whereas heavy coffee-drinkers with rs4998386_TC genotype had 59% lower risk (P = 6×10(-13)). Imputation revealed a block of SNPs that achieved P(2df)<5×10(-8) in GWAIS, and OR = 0.41, P = 3×10(-8) in heavy coffee-drinkers. This study is proof of concept that inclusion of environmental factors can help identify genes that are missed in GWAS. Both adenosine antagonists (caffeine-like) and glutamate antagonists (GRIN2A-related) are being tested in clinical trials for treatment of PD. GRIN2A may be a useful pharmacogenetic marker for subdividing individuals in clinical trials to determine which medications might work best for which patients.
Collapse
Affiliation(s)
- Taye H. Hamza
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | - Honglei Chen
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Erin M. Hill-Burns
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | - Shannon L. Rhodes
- Department of Epidemiology, School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Montimurro
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | - Denise M. Kay
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | - Albert Tenesa
- Institute of Genetics and Molecular Medicine and The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria I. Kusel
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | - Patricia Sheehan
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| | | | - Dora Yearout
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Ali Samii
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - John W. Roberts
- Virginia Mason Medical Center, Seattle, Washington, United States of America
| | - Pinky Agarwal
- Booth Gardner Parkinson's Care Center, Evergreen Hospital Medical Center, Kirkland, Washington, United States of America
| | - Yvette Bordelon
- Department of Neurology, School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Yikyung Park
- Nutritional Epidemiology Branch, Divisions of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Liyong Wang
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jianjun Gao
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Jeffery M. Vance
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Silviu-Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - William K. Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Beate Ritz
- Department of Epidemiology, School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurology, School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Environmental Health Sciences, Center for Occupational and Environmental Health, School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - John Nutt
- Department of Neurology, Oregon Health and Sciences University, Portland, Oregon, United States of America
| | - Stewart A. Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Cyrus P. Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Haydeh Payami
- New York State Department of Health Wadsworth Center, Albany, New York, United States of America
| |
Collapse
|
18
|
Armentero MT, Pinna A, Ferré S, Lanciego JL, Müller CE, Franco R. Past, present and future of A(2A) adenosine receptor antagonists in the therapy of Parkinson's disease. Pharmacol Ther 2011; 132:280-99. [PMID: 21810444 DOI: 10.1016/j.pharmthera.2011.07.004] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/07/2011] [Indexed: 12/20/2022]
Abstract
Several selective antagonists for adenosine A(2A) receptors (A(2A)R) are currently under evaluation in clinical trials (phases I to III) to treat Parkinson's disease, and they will probably soon reach the market. The usefulness of these antagonists has been deduced from studies demonstrating functional interactions between dopamine D₂ and adenosine A(2A) receptors in the basal ganglia. At present it is believed that A(2A)R antagonists can be used in combination with the dopamine precursor L-DOPA to minimize the motor symptoms of Parkinson's patients. However, a considerable body of data indicates that in addition to ameliorating motor symptoms, adenosine A(2A)R antagonists may also prevent neurodegeneration. Despite these promising indications, one further issue must be considered in order to develop fully optimized antiparkinsonian drug therapy, namely the existence of (hetero)dimers/oligomers of G protein-coupled receptors, a topic that is currently the focus of intense debate within the scientific community. Dopamine D₂ receptors (D₂Rs) expressed in the striatum are known to form heteromers with A(2A) adenosine receptors. Thus, the development of heteromer-specific A(2A) receptor antagonists represents a promising strategy for the identification of more selective and safer drugs.
Collapse
Affiliation(s)
- Marie Therese Armentero
- Laboratory of Functional Neurochemistry, Interdepartmental Research Centre for Parkinson's Disease, IRCCS National Institute of Neurology "C. Mondino", Pavia, Italy
| | | | | | | | | | | |
Collapse
|
19
|
Black YD, Xiao D, Pellegrino D, Kachroo A, Brownell AL, Schwarzschild MA. Protective effect of metabotropic glutamate mGluR5 receptor elimination in a 6-hydroxydopamine model of Parkinson's disease. Neurosci Lett 2010; 486:161-5. [PMID: 20854878 DOI: 10.1016/j.neulet.2010.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 09/12/2010] [Accepted: 09/14/2010] [Indexed: 11/26/2022]
Abstract
Pharmacologic or genetic blockade of metabotropic glutamate mGlu5 receptors (mGluR5) has been shown to attenuate parkinsonian motor deficits and protect nigrostriatal neurons from damage in the acute MPTP model of Parkinson's disease (PD), suggesting that therapeutically targeting the mGluR5 receptor may offer a novel approach to improving motor symptoms and/or slowing neurodegeneration in PD. This study further explored the neuroprotective potential of targeting mGluR5 receptors. We examined the behavioral and neurochemical effects of receptor elimination on toxicity induced by intra-striatal application of 6-hydroxydopamine (6-OHDA), thought to represent a comparatively progressive model of PD. mGluR5 knockout (KO) mice and wild-type (WT) littermates received unilateral 6-OHDA infusions. Reflecting the imbalance expected following unilateral infusion, WT but not KO mice demonstrated predominantly ipsilateral forepaw use and robust ipsilateral amphetamine-induced rotation. Further, performance on the vertical pole descent task was profoundly impaired in WT mice, while KO mice completed the task significantly faster. Consistent with the behavioral observations, neurochemical analyses of striatal dopamine depletion showed significantly diminished severity in KO mice with only 64% of striatal dopamine lost, compared to 92% in WT mice. The absence of brain mGluR5 receptors in living KO mice was verified using positron emission tomography (PET). Our findings substantiate the key role of mGluR5 receptors in animal models of PD, strengthening the rationale for the development of mGluR5 antagonists for their neuroprotective, as well as symptomatic, benefit.
Collapse
Affiliation(s)
- Yolanda D Black
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
20
|
Leaver KR, Allbutt HN, Creber NJ, Kassiou M, Henderson JM. Neuroprotective effects of a selective N-methyl-D-aspartate NR2B receptor antagonist in the 6-hydroxydopamine rat model of Parkinson's disease. Clin Exp Pharmacol Physiol 2009; 35:1388-94. [PMID: 18785982 DOI: 10.1111/j.1440-1681.2008.05046.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Current pharmacotherapies for the treatment of Parkinson's disease (PD) are largely symptomatic and do not attenuate the characteristic nigral (dopamine) cell loss. 2. Using the 6-hydroxydopamine (6-OHDA) rat model of PD, we investigated the novel, potentially neuroprotective compound BZAD-01, which is an N-methyl-D-aspartate (NMDA) glutamate receptor antagonist selective for the NR2B subunit. 3. Forty female Sprague-Dawley rats were pretreated with either 10 mg/kg BZAD-01 or vehicle (5% sucrose and 0.1% ascorbate) in their drinking water for 11 days prior to and for 3 days following 6-OHDA surgery. During surgery, rats received an injection of either a toxic dose of 16 microg 6-OHDA or a non-toxic dose of 1 microg 6-OHDA (sham) into the medial forebrain bundle. A series of behavioural tests, including curling (measuring body axis bias), head position bias and narrow beam, was performed fortnightly for 8 weeks after surgery to assess the effects of BZAD-01 pretreatment on parkinsonism. Drug-induced rotational asymmetry was also assessed just before rats were killed. Post-mortem immunohistochemistry was performed to quantify the degree of nigral dopamine cell loss. 4. Pretreatment of 6-OHDA-lesioned rats with BZAD-01 significantly reduced the amount of dopamine cell loss and significantly improved all behavioural measures. Furthermore, there was no significant difference in any of the behavioural measures between lesioned rats pretreated with BZAD-01 and rats that underwent sham surgery.
Collapse
Affiliation(s)
- K R Leaver
- Department of Pharmacology, Bosch Institute and School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|
21
|
|
22
|
Luquin N, Mitrofanis J. Does the cerebral cortex exacerbate dopaminergic cell death in the substantia nigra of 6OHDA-lesioned rats? Parkinsonism Relat Disord 2008; 14:213-23. [DOI: 10.1016/j.parkreldis.2007.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 06/25/2007] [Accepted: 08/13/2007] [Indexed: 10/22/2022]
|
23
|
Blandini F, Levandis G, Bazzini E, Nappi G, Armentero MT. Time-course of nigrostriatal damage, basal ganglia metabolic changes and behavioural alterations following intrastriatal injection of 6-hydroxydopamine in the rat: new clues from an old model. Eur J Neurosci 2007; 25:397-405. [PMID: 17284180 DOI: 10.1111/j.1460-9568.2006.05285.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite the progressive development of innovative animal models for Parkinson's disease, the intracerebral infusion of neurotoxin 6-hydroxydopamine (6-OHDA) remains the most widely used means to induce an experimental lesion of the nigrostriatal pathway in the animal, due to its relatively low complexity and cost, coupled with the high reproducibility of the lesion obtained. To gain new information from such a classic model, we studied the time-course of the nigrostriatal damage, metabolic changes in the basal ganglia nuclei (cytochrome oxidase activity) and behavioural modifications (rotational response to apomorphine) following unilateral injection of 6-OHDA into the corpus striatum of rat, over a 4-week period. Striatal infusion of 6-OHDA caused early damage of dopaminergic terminals, followed by a slowly evolving loss of dopaminergic cell bodies in the substantia nigra pars compacta, which became apparent during the second week post-injection and peaked at the 28th day post-infusion; the rotational response to apomorphine was already present at the first time point considered (Day 1), and remained substantially stable throughout the 4-week period of observation. The evolution of the nigrostriatal lesion was accompanied by complex changes in the metabolic activity of the other basal ganglia nuclei investigated (substantia nigra pars reticulata, entopeduncular nucleus, globus pallidus and subthalamic nucleus), which led, ultimately, to a generalized, metabolic hyperactivity, ipsilaterally to the lesion. However, peculiar patterns of metabolic activation, or inhibition, characterized the post-lesional responses of each nucleus, in the early and intermediate phases, with peculiar response profiles that varied closely related to the functional position occupied within the basal ganglia circuitry.
Collapse
Affiliation(s)
- Fabio Blandini
- Laboratory of Functional Neurochemistry, IRCCS Neurological Institute C. Mondino, Pavia, Italy.
| | | | | | | | | |
Collapse
|
24
|
Vernon AC, Zbarsky V, Datla KP, Dexter DT, Croucher MJ. Selective activation of group III metabotropic glutamate receptors by L-(+)-2-amino-4-phosphonobutryic acid protects the nigrostriatal system against 6-hydroxydopamine toxicity in vivo. J Pharmacol Exp Ther 2007; 320:397-409. [PMID: 17012606 DOI: 10.1124/jpet.106.108159] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Evidence from several studies suggests that the progressive degeneration of dopaminergic (DA) neurones of the substantia nigra pars compacta (SNc) in Parkinson's disease (PD) may in part be due to excessive release of glutamate from subthalamic projections onto nigral DA neurones. Previous in vitro studies have demonstrated that selective activation of Group III metabotropic glutamate receptors (mGluR) negatively modulates excitatory transmission in the SNc and is neuroprotective against glutamate-mediated toxicity. Consistent with this, we have reported preliminary data indicating that the selective group III mGluR agonist l-(+)-2-amino-4-phosphonobutyric acid (l-AP4) can also protect the nigrostriatal system against 6-hydroxydopamine (6-OHDA) toxicity in vivo. We have now extended these preliminary studies in this model and report here that both acute and subchronic intranigral injections of l-AP4 provide significant protection of the nigrostriatal system against 6-OHDA toxicity. This neuroprotection displays a bell-shaped profile with a clear concentration-dependent relationship. In contrast, when administered to animals 7 days post-6-OHDA lesioning, l-AP4 significantly protects the functionality but not the integrity of the nigrostriatal system. We further demonstrate that neuroprotection by l-AP4 in vivo is reversed by coadministration of the selective Group III mGluR antagonist (RS)-alpha-methylserine-O-phosphate, confirming a receptor-mediated mechanism of action. These data provide further compelling evidence that selective activation of Group III mGluR is neuroprotective in an in vivo experimental model of PD, a finding that may have important implications for the future treatment of this disease.
Collapse
Affiliation(s)
- Anthony C Vernon
- Faculty of Medicine, Sir Alexander Fleming Building, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | |
Collapse
|
25
|
|
26
|
Bonsi P, Cuomo D, Picconi B, Sciamanna G, Tscherter A, Tolu M, Bernardi G, Calabresi P, Pisani A. Striatal metabotropic glutamate receptors as a target for pharmacotherapy in Parkinson’s disease. Amino Acids 2006; 32:189-95. [PMID: 16715415 DOI: 10.1007/s00726-006-0320-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 01/12/2006] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopamine (DA)-containing neurons in the substantia nigra pars compacta (SNc). The symptoms are resting tremor, slowness of movement, rigidity and postural instability. Evidence that an imbalance between dopaminergic and cholinergic transmission takes place within the striatum led to the utilization of DA precursors, DA receptor agonists and anticholinergic drugs in the symptomatic therapy of PD. However, upon disease progression the therapy becomes less effective and debilitating effects such as dyskinesias and motor fluctuations appear. Hence, the need for the development of alternative therapeutic strategies has emerged. Several observations in different experimental models of PD suggest that blockade of excitatory amino acid transmission exerts antiparkinsonian effects. In particular, recent studies have focused on metabotropic glutamate receptors (mGluRs). Drugs acting on group I and II mGluRs have indeed been proven useful in ameliorating the parkinsonian symptoms in animal models of PD and therefore might represent promising therapeutic targets. This beneficial effect could be due to the reduction of both glutamatergic and cholinergic transmission. A novel target for drugs acting on mGluRs in PD therapy might be represented by striatal cholinergic interneurons. Indeed, the activation of mGluR2, highly expressed on this cell type, is able to reduce calcium-dependent plateau potentials by interfering with somato-dendritic N-type calcium channel activity, in turn reducing ACh release in the striatum. Similarly, the blockade of both group I mGluR subtypes reduces cholinergic interneuron excitability, and decreases striatal ACh release. Thus, targeting mGluRs located onto cholinergic interneurons might result in a beneficial pharmacological effect in the parkinsonian state.
Collapse
Affiliation(s)
- P Bonsi
- Laboratorio di Neurofisiologia, I.R.C.C.S. Fondazione Santa Lucia - C.E.R.C., European Brain Research Institute, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Betarbet R, Sherer TB, Di Monte DA, Greenamyre JT. Mechanistic approaches to Parkinson's disease pathogenesis. Brain Pathol 2006; 12:499-510. [PMID: 12408237 PMCID: PMC8095781 DOI: 10.1111/j.1750-3639.2002.tb00468.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disorder marked by nigrostriatal dopaminergic degeneration and development of cytoplasmic proteinaceous aggregates known as Lewy bodies. Although the pathogenic mechanisms responsible for PD are not completely understood, many clues have come from biochemical, epidemiological, and genetic studies. Mutations in certain genes found in rare, familial cases of PD, such as alpha-synuclein and parkin, suggest a role for the ubiquitin-proteosome system and aberrant protein aggregation. Biochemical analyses have implicated mitochondrial dysfunction in PD. Epidemiological and animal model studies point to a role for environmental toxins, some of which are mitochondrial inhibitors. Mitochondrial dysfunction, resulting from either genetic defects, environmental exposures or an interaction between the two, may cause alpha-synuclein aggregation or neurodegeneration through oxidative stress or excitotoxicity. A better understanding of the mechanisms underlying PD should reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Ranjita Betarbet
- Center for Neurodegenerative Disease and Department of Neurology, Emory University, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
28
|
Armentero MT, Fancellu R, Nappi G, Bramanti P, Blandini F. Prolonged blockade of NMDA or mGluR5 glutamate receptors reduces nigrostriatal degeneration while inducing selective metabolic changes in the basal ganglia circuitry in a rodent model of Parkinson's disease. Neurobiol Dis 2006; 22:1-9. [PMID: 16289868 DOI: 10.1016/j.nbd.2005.09.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 09/19/2005] [Accepted: 09/27/2005] [Indexed: 11/21/2022] Open
Abstract
We compared the neuroprotective and metabolic effects of chronic treatment with ionotropic or metabotropic glutamate receptor antagonists, in rats bearing a unilateral nigrostriatal lesion induced by 6-hydroxydopamine (6-OHDA). The ionotropic, N-methyl-D-aspartate receptor antagonist MK-801 increased cell survival in the substantia nigra pars compacta (SNc) and corrected the metabolic hyperactivity (increased cytochrome oxidase activity) of the ipsilateral substantia nigra pars reticulata (SNr) associated with the lesion, but showed no effects on the 6-OHDA-induced hyperactivity of the subthalamic nucleus (STN). Significant-although less pronounced-protection of SNc neurons was also observed following treatment with the metabotropic glutamate receptor (mGluR5) antagonist 2-methyl-6-(phenylehtynyl)-pyridine (MPEP). As opposed to MK-801, MPEP abolished the STN metabolic hyperactivity associated with the nigrostriatal lesion, without affecting SNr activity. Specific modulation of STN hyperactivity obtained with mGluR5 blockade may, therefore, open interesting perspectives for the use of this class of compounds in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Marie-Thérèse Armentero
- Laboratory of Functional Neurochemistry, IRCCS Neurological Institute "C. Mondino", Via Mondino, 2, 27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
29
|
Winter C, Hosmann K, Harnack D, Meissner W, Paul G, Morgenstern R, Kupsch A. Subthalamic nucleus lesioning inhibits expression and phosphorylation of c-Jun in nigral neurons in the rat's 6-OHDA model of Parkinson's disease. Synapse 2006; 60:69-80. [PMID: 16598703 DOI: 10.1002/syn.20269] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Parkinson's Disease (PD) is characterized by a loss of nigral dopamine (DA) neurons, followed by a striatal DA deficit. Inhibition of the subthalamic nucleus (STN) reverses L-DOPA sensitive motor symptoms and improves efficacy of pharmacotherapy in PD-patients. The underlying mechanism of these effects, however, remains largely unknown. Previously, we could show in the rat's 6-hydroxyDA (6-OHDA) model of PD that ablative STN-lesioning exerts functionally neuroprotective effects on the DAergic nigrostriatal pathway against 6-OHDA toxicity, in terms of elevating the number of tyrosine hydroxylase (TH)-expressing neurons rather than enhancing the total number of cells surviving 2 and 6 weeks post lesioning, as assessed via fluorogold staining. These data were correlated with increased functional recovery of 6-OHDA-lesioned rats with preceding STN-lesioning. Here, we extend the previous study design to observation periods of up to 12 weeks to assess long-term effects. Furthermore, to elucidate cellular mechanisms underlying potential neuroprotective effects, we explore the regulation of cellular markers involved in neurodegenerative cascades via immunocytochemistry. We show that preceding STN-lesioning significantly inhibits 6-OHDA induced expression/phosphorylation of the transcription factor c-Jun in surviving nigral neurons in comparison with controls. However, we also demonstrate that functionally neuroprotective effects of preceding STN-lesioning subside after 12 weeks, as assessed with TH immunostaining. We therefore conclude that c-Jun induction/phosphorylation is involved in 6-OHDA toxicity and that STN-lesioning transiently preserves of dopaminergic phenotype of nigral neurons partially via delaying the induction and attenuating the expression and phosphorylation of c-Jun.
Collapse
Affiliation(s)
- Christine Winter
- Department of Neurology, Charité-University Medicine Berlin, Campus Charité Mitte, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
Conn PJ, Battaglia G, Marino MJ, Nicoletti F. Metabotropic glutamate receptors in the basal ganglia motor circuit. Nat Rev Neurosci 2005; 6:787-98. [PMID: 16276355 DOI: 10.1038/nrn1763] [Citation(s) in RCA: 250] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In recent years there have been tremendous advances in our understanding of the circuitry of the basal ganglia and our ability to predict the behavioural effects of specific cellular changes in this circuit on voluntary movement. These advances, combined with a new understanding of the rich distribution and diverse physiological roles of metabotropic glutamate receptors in the basal ganglia, indicate that these receptors might have a key role in motor control and raise the exciting possibility that they might provide therapeutic targets for the treatment of Parkinson's disease and related disorders.
Collapse
Affiliation(s)
- P Jeffrey Conn
- Program in Translational Neuropharmacology, Department of Pharmacology, Vanderbilt University Medical Center, 23rd Avenue South at Pierce, 417-D Preson Research Building, Nashville, Tennessee 37232-6600, USA.
| | | | | | | |
Collapse
|
31
|
Sousa SC, Castilho RF. Protective effect of melatonin on rotenone plus Ca2+-induced mitochondrial oxidative stress and PC12 cell death. Antioxid Redox Signal 2005; 7:1110-6. [PMID: 16115015 DOI: 10.1089/ars.2005.7.1110] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic systemic inhibition of mitochondrial respiratory chain complex I by rotenone causes nigrostriatal dopaminergic degeneration in rats, producing an in vivo experimental model of Parkinson's disease. We recently showed that micromolar Ca2+ concentrations strongly stimulate the release of reactive oxygen species in rotenone-treated isolated rat brain mitochondria. In the present work, we show that the natural antioxidant melatonin inhibits Ca2+ plus rotenone-induced oxidative stress in isolated rat brain mitochondria. In addition, the Ca2+ ionophore A23187 strongly potentiates rotenone-induced death of intact cultured pheochromocytoma (PC12) cells, in a mechanism sensitive to melatonin. Moreover, melatonin inhibits the detection of reactive oxygen species release in PC12 cells treated with rotenone plus A23187. Melatonin does not alter free Ca2+ concentrations or the inhibitory effect of rotenone on mitochondrial complex I. We conclude that micromolar Ca2+ concentrations stimulate neuronal cell death induced by mitochondrial complex I inhibition in a mechanism involving oxidative stress, preventable by the antioxidant melatonin.
Collapse
Affiliation(s)
- Solange C Sousa
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, Brazil
| | | |
Collapse
|
32
|
Kress GJ, Reynolds IJ. Dopaminergic neurotoxins require excitotoxic stimulation in organotypic cultures. Neurobiol Dis 2005; 20:639-45. [PMID: 15996475 DOI: 10.1016/j.nbd.2005.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2004] [Revised: 04/09/2005] [Accepted: 04/26/2005] [Indexed: 10/25/2022] Open
Abstract
We have investigated the properties of the dopaminergic neurotoxins 6-hydroxydopamine, 1-methyl-4-phenylpyridinium and rotenone using an organotypic culture that included slices of substantia nigra, striatum and cortex maintained for about 20 days in vitro. At this age, the organotypic culture contains dopaminergic neurons, visualized using tyrosine hydroxylase (TH) immunohistochemistry, that project into the striatal slice and extend up to 1 mm into the cortical slice. Using TH immunohistochemistry to assess survival of dopaminergic neurons, we found that the three dopaminergic toxins alone were not selectively neurotoxic. However, the addition of a low concentration of N-methyl-d-aspartate together with each individual toxin resulted in profound injury to the dopaminergic neurons, reflected by the loss of cell bodies and the fragmentation of processes. The combined toxicity was completely blocked by MK801. To assess the specificity of the injury, we measured the diameter of cell nuclei in the organotypic culture stained with Hoechst 33342 because the nucleus shrinks when neurons are injured. These measurements showed that the combined toxin treatment selectively injured only the TH immunoreactive cells. Thus, in a model culture system where dopaminergic neurons innervate appropriate targets, excitotoxicity appears to be essential for the manifestation of the toxic actions of 6-hydroxydopamine, 1-methyl-4-phenylpyridinium and rotenone.
Collapse
Affiliation(s)
- Geraldine J Kress
- Department of Pharmacology, University of Pittsburgh, W1351 Biomedical Science Tower, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
33
|
Bové J, Serrats J, Mengod G, Cortés R, Tolosa E, Marin C. Neuroprotection induced by the adenosine A2A antagonist CSC in the 6-OHDA rat model of parkinsonism: effect on the activity of striatal output pathways. Exp Brain Res 2005; 165:362-74. [PMID: 15968457 DOI: 10.1007/s00221-005-2302-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2004] [Accepted: 02/02/2005] [Indexed: 12/18/2022]
Abstract
In Parkinson's disease (PD), the striatal dopamine depletion and the following overactivation of the indirect pathway of the basal ganglia leads to very early disinhibition of the subthalamic nucleus (STN) that may contribute to the progression of PD by glutamatergic overstimulation of the dopaminergic neurons in the substantia nigra. Adenosine A2A antagonism has been demonstrated to attenuate the overactivity of the striatopallidal pathway. To investigate whether neuroprotection exerted by the A2A antagonist 8-(3-chlorostyryl)caffeine (CSC) correlates with a diminution of the striatopallidal pathway activity, we have examined the changes in the mRNA encoding for enkephalin, dynorphin, and adenosine A2A receptors by in situ hybridization induced by subacute systemic pretreatment with CSC in rats with striatal 6-hydroxydopamine(6-OHDA) administration. Animals received CSC for 7 days until 30 min before 6-OHDA intrastriatal administration. Vehicle-treated group received a solution of dimethyl sulfoxide. CSC pretreatment partially attenuated the decrease in nigral tyrosine hydroxylase immunoreactivity induced by 6-OHDA, whereas no modification of the increase in preproenkephalin mRNA expression in the dorsolateral striatum was observed. The neuroprotective effect of the adenosine A2A antagonist CSC in striatal 6-OHDA-lesioned rats does not result from a normalization of the increase in striatal PPE mRNA expression in the DL striatum, suggesting that other different mechanisms may be involved.
Collapse
Affiliation(s)
- Jordi Bové
- Laboratori de Neurologia Experimental, Area de Neurociències, Fundació Clinic-Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel 170, 08036 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Valenti O, Mannaioni G, Seabrook GR, Conn PJ, Marino MJ. Group III metabotropic glutamate-receptor-mediated modulation of excitatory transmission in rodent substantia nigra pars compacta dopamine neurons. J Pharmacol Exp Ther 2005; 313:1296-304. [PMID: 15761115 DOI: 10.1124/jpet.104.080481] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glutamate plays an important role in the regulation of dopamine neuron activity. In particular, the glutamatergic input from the subthalamic nucleus is thought to provide control over dopamine neuron firing patterns. The degeneration of dopamine neurons in the substantia nigra pars compacta (SNc) observed in Parkinson's disease (PD) is believed to be due to a complex interplay of factors, including oxidative stress and mitochondrial dysfunction. Although glutamate is not the primary cause of cell death in PD, there is evidence suggesting excessive glutamate release onto dopamine neurons may play a role in continued degeneration. Although many studies have focused on the role of glutamate in the SNc, little work has been directed at exploring the modulatory control of glutamate release in this region. Previous studies have found a high-potency inhibitory effect of nonselective group III mGluR agonist on glutamatergic transmission in the SNc. Using whole-cell patch-clamp methods and novel pharmacological tools, we have determined that mGluR4 mediates the group III mGluR modulation of excitatory transmission in the rat SNc. The group III mGluR-selective agonist l-(+)-2-amino-4-phosphonobutyric acid inhibits excitatory transmission in the SNc at low micromolar concentrations with a maximal inhibition occurring at 3 muM. This effect was potentiated by the mGluR4-selective allosteric modulator N-phenyl-7-(hydroxymino)cyclopropa[b]chromen-1a-carboxamide and was not mimicked by the mGluR8-selective agonist (S)-3,4-dicarboxyphenylglycine. Interestingly, in an attempt to employ knockout mice to confirm the role of mGluR4, we discovered an apparent species difference suggesting that in mice, both mGluR4 and mGluR8 modulate excitatory transmission in the SNc.
Collapse
Affiliation(s)
- Ornella Valenti
- Neuroscience Drug Discovery, Movement Disorders, Merck Research Laboratories, West Point, PA 19486, USA
| | | | | | | | | |
Collapse
|
35
|
Shen KZ, Johnson SW. Dopamine depletion alters responses to glutamate and GABA in the rat subthalamic nucleus. Neuroreport 2005; 16:171-4. [PMID: 15671871 DOI: 10.1097/00001756-200502080-00021] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We used whole-cell recordings to compare currents evoked by glutamate and GABA receptor agonists in subthalamic nucleus neurons located ipsilateral and contralateral to unilateral 6-hydroxydopamine (6-OHDA) injections into the substantia nigra zona compacta. The ratio of currents evoked by AMPA (0.6 microM) and NMDA (20 microM) was significantly greater in neurons recorded ipsilateral to 6-OHDA lesions compared with the ratio of currents recorded in control (contralateral) neurons. Both the GABA(A) agonist isoguvacine (20 microM) and the GABA(B) agonist baclofen (10 microM) evoked significantly greater outward currents in subthalamic nucleus neurons ipsilateral to the lesion compared to contralateral neurons. We conclude that chronic dopamine depletion up-regulates expression of GABA receptors and shifts the functional expression of ionotropic glutamate receptor subtype from NMDA to AMPA receptors in subthalamic nucleus.
Collapse
Affiliation(s)
- Ke-Zhong Shen
- 1Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|
36
|
Wang XJ, Xu JX. Possible involvement of Ca2+ signaling in rotenone-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neurosci Lett 2005; 376:127-32. [PMID: 15698934 DOI: 10.1016/j.neulet.2004.11.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 11/14/2004] [Accepted: 11/16/2004] [Indexed: 11/18/2022]
Abstract
Rotenone, an inhibitor of mitochondrial respiratory chain complex I, is a useful tool to elicit animal model of Parkinson's disease. Rotenone-induced neuronal apoptosis may contribute to the etiology of Parkinson's disease. However, the mechanism of rotenone-induced apoptosis is not fully understood. In the present study, we show that Ca2+ signaling is essential for rotenone-induced apoptosis in human neuroblastoma SH-SY5Y cells. By using Fluo-3/AM and Fura-2/AM, the fluorescent calcium indicator, rotenone was found to cause a rise in intracellular free Ca2+ ([Ca2+]i). The intracellular Ca2+ chelator BAPTA attenuated rotenone-induced apoptosis. Notably, Ca2+ suppression also prevented rotenone-induced apoptotic related events including reactive oxygen species production, G2/M cell cycle arrest and caspase activation, suggesting that Ca2+ signaling is upstream to these events. In the absence of extracellular Ca2+, the rotenone-induced [Ca2+]i elevation was inhibited. Further, the voltage-dependent Ca2+ channel blocker nifedipine suppressed most of the elevation of [Ca2+]i induced by rotenone. These results demonstrate that rotenone leads to an elevation in [Ca2+]i through Ca2+ influx by the opening of voltage-gated Ca2+ channel. This study of rotenone may help to elucidate the neurodegenerative mechanims in Parkinson's disease.
Collapse
Affiliation(s)
- Xin-Jian Wang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | | |
Collapse
|
37
|
Marin C, Bové J, Serrats J, Cortés R, Mengod G, Tolosa E. The kappa opioid agonist U50,488 potentiates 6-hydroxydopamine-induced neurotoxicity on dopaminergic neurons. Exp Neurol 2005; 191:41-52. [PMID: 15589511 DOI: 10.1016/j.expneurol.2004.08.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Revised: 08/16/2004] [Accepted: 08/19/2004] [Indexed: 11/26/2022]
Abstract
Several observations support the hypothesis that kappa opioid (kappa-opioid) receptor agonism may contribute to neurotoxicity, but other reports have suggested that certain kappa-agonists can attenuate neurological dysfunction. Degeneration of dopaminergic neurons in the substantia nigra is the pathological hallmark of Parkinson's disease. Therefore, it is of particular interest to study whether kappa-opioid receptor agonism has an influence on the progressive degeneration of dopaminergic neurons. We have investigated the effect exerted by the selective kappa-agonist U50,488 on the neurotoxicity induced by intrastriatal 6-hydroxydopamine (6-OHDA) administration on dopaminergic neurons. Male Sprague-Dawley rats received an acute (0.5 mg/kg) or subacute (0.5 mg/kg, twice at day, for 7 days) administration of U50,488, receiving the last dose 30 min before intrastriatal 6-OHDA administration. Acute or subacute U50,488 pretreatment potentiated the 6-OHDA-induced decrease in the number of nigral tyrosine hydroxylase immunoreactive neurons (P < 0.05). Acute U50,488 pretreated animals showed a tendency, although not statistically significant to increase striatal mRNA encoding for enkephalin (PPE mRNA). Subacute U50,488 significantly potentiated the increase in PPE mRNA induced by 6-OHDA (P < 0.05). The present results show a neurotoxic effect of the kappa agonist U50,488 on dopaminergic neurons in rats with a striatal lesion induced by 6-OHDA. This neurotoxic effect is associated to an increase in striatal PPE mRNA levels, suggesting that an increase in the indirect pathway activity and consequently an increase in the activity of the subthalamo-nigral pathway might be involved in this phenomenon.
Collapse
Affiliation(s)
- C Marin
- Laboratori de Neurologia Experimental, Area de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
38
|
Domenici MR, Potenza RL, Martire A, Coccurello R, Pèzzola A, Reggio R, Tebano MT, Popoli P. Chronic treatment with the mGlu5R antagonist MPEP reduces the functional effects of the mGlu5R agonist CHPG in the striatum of 6-hydroxydopamine-lesioned rats: Possible relevance to the effects of mGlu5R blockade in Parkinson's disease. J Neurosci Res 2005; 80:646-54. [PMID: 15880742 DOI: 10.1002/jnr.20489] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This study was designed to test whether chronic treatment with the metabotropic glutamate receptor 5 (mGlu5R) antagonist MPEP showed antiparkinsonian effects in rats unilaterally lesioned with 6-hydroxydopamine (6-OHDA) (a "classic" model of Parkinson's disease, PD), and to evaluate whether chronic MPEP influenced the functional properties and/or the expression of striatal mGlu5Rs. Wistar rats were lesioned with 6-OHDA and then treated with MPEP (3 mg/kg/day, i.p.) or its vehicle over 2 weeks. Chronic MPEP did not induce measurable antiparkinsonian effects, since no differences were found between MPEP- and vehicle-treated animals in the pattern of L-DOPA-induced contralateral rotations. In corticostriatal slices taken from animals chronically treated with MPEP, the functional effects of the mGlu5R agonist CHPG were significantly reduced in the lesioned vs. the intact side, while no changes were found in slices taken from vehicle-treated rats. The binding of [3H]MPEP to striatal membranes showed that neither the maximal number of binding sites (Bmax) nor the dissociation constant (Kd) were changed by the lesion and/or by chronic MPEP. While chronic MPEP did not potentiate L-DOPA-induced turning in a classical model of PD, its ability to reduce mGlu5R-associated signal could help to explain the neuroprotective/antiparkinsonian effects observed in other models of PD.
Collapse
|
39
|
Srinivasan J, Schmidt WJ. Treatment with alpha2-adrenoceptor antagonist, 2-methoxy idazoxan, protects 6-hydroxydopamine-induced Parkinsonian symptoms in rats: neurochemical and behavioral evidence. Behav Brain Res 2004; 154:353-63. [PMID: 15313023 DOI: 10.1016/j.bbr.2004.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Revised: 03/02/2004] [Accepted: 03/04/2004] [Indexed: 11/22/2022]
Abstract
Noradrenaline, not only functions as a synaptic transmitter, but also promotes neural differentiation and regenerative processes. In Parkinson's disease, besides the dopaminergic degeneration, noradrenergic neurons of locus coeruleus origin degenerate as well. Drugs enhancing noradrenergic transmission in the locus coeruleus (e.g. alpha2-adrenoceptor antagonists) have been shown to be neuroprotective against Huntington's and ischemic animal models. However, in Parkinsonian animal models, most of the studies evaluated the worsening of experimental nigral neurodegeneration after locus coeruleus lesions. Here, it has been tested, whether treatment with the selective alpha2-adrenoceptor antagonist, 2-methoxy idazoxan (2.5 mg/kg i.p., twice daily for 5 days), before an experimental lesion to nigra, protects dopaminergic neurodegeneration. Dopaminergic degeneration was produced by 6-hydroxydopamine lesion in the median forebrain bundle. The concentrations of dopamine, 5-hydroxytryptamine and its metabolites were analysed in the various regions of the basal ganglia. The concentrations of noradrenaline and dopamine were measured in the regions innervated by locus coeruleus neurons and in the basal ganglia respectively, after 2-methoxy idazoxan treatment. The Parkinsonian behavior was assessed by catalepsy and activity test. 2-Methoxy idazoxan specifically increased the concentration of noradrenaline in the brain regions, innervated by locus coeruleus neurons. 6-OHDA lesion strongly depleted the concentration of dopamine and its metabolites in the striatum and SN, producing catalepsy and hypoactivity. Multiple treatments with 2-methoxy idazoxan reduced some of the observed neurochemical and behavioral indices of 6-hydroxydopamine-induced Parkinsonism, indicating neuroprotection. Although the mechanism underlying the neuroprotective property remains elusive, the therapeutic usage of alpha2-antagonists might be helpful in slowing the neuronal death and progression of Parkinson's disease.
Collapse
Affiliation(s)
- J Srinivasan
- Neuropharmacology, Zoological Institute, University of Tuebingen, Auf der Morgenstelle 28E, 72076 Tuebingen, Germany
| | | |
Collapse
|
40
|
Krishnamurthi R, Stott S, Maingay M, Faull RLM, McCarthy D, Gluckman P, Guan J. N-terminal tripeptide of IGF-1 improves functional deficits after 6-OHDA lesion in rats. Neuroreport 2004; 15:1601-4. [PMID: 15232291 DOI: 10.1097/01.wnr.0000127461.15985.07] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Central administration of N-terminal tripeptide of IGF-1 (GPE) prevents the loss of dopamine neurons. We now examine effects of GPE administered peripherally, on long-term functional recovery after 6-OHDA lesion in rats. GPE treatment (3 mg/kg, i.p.), 3 days after the lesion reduced the number of rotations (p<0.005) and the time over meter (p<0.005) compared to vehicle treatment. Step length and number of adjusting steps were increased in the GPE group (p<0.005), particularly at 12 weeks post lesion. However, GPE treatment did not prevent the loss of tyrosine hydroxylase in the substantia nigra pars compacta and the striatum. The study suggests that peripheral administration of GPE after onset of nigrostriatal dopamine depletion improves long-term Parkinsonian motor deficits, independent of neuronal outcome.
Collapse
Affiliation(s)
- Rita Krishnamurthi
- Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
41
|
Marino MJ, Valenti O, Conn PJ. Glutamate receptors and Parkinson's disease: opportunities for intervention. Drugs Aging 2004; 20:377-97. [PMID: 12696997 DOI: 10.2165/00002512-200320050-00006] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Parkinson's disease is a debilitating neurodegenerative movement disorder that is the result of a degeneration of dopaminergic neurons in the substantia nigra pars compacta. The resulting loss of striatal dopaminergic tone is believed to underlie a series of changes in the circuitry of the basal ganglia that ultimately lead to severe motor disturbances due to excessive basal ganglia outflow. Glutamate plays a central role in the disruption of normal basal ganglia function, and it has been hypothesised that agents acting to restore normal glutamatergic function may provide therapeutic interventions that bypass the severe motor side effects associated with current dopamine replacement strategies. Analysis of the effects of glutamate receptor ligands in the basal ganglia circuit suggests that both ionotropic and metabotropic glutamate receptors could have antiparkinsonian actions. In particular, NMDA receptor antagonists that selectively target the NR2B subunit and antagonists of the metabotropic glutamate receptor mGluR5 appear to hold promise and deserve future attention.
Collapse
Affiliation(s)
- Michael J Marino
- Department of Neuroscience, Merck Research Laboratories, West Point, Pennsylvania 19486-0004, USA
| | | | | |
Collapse
|
42
|
Paul G, Meissner W, Rein S, Harnack D, Winter C, Hosmann K, Morgenstern R, Kupsch A. Ablation of the subthalamic nucleus protects dopaminergic phenotype but not cell survival in a rat model of Parkinson's disease. Exp Neurol 2004; 185:272-80. [PMID: 14736508 DOI: 10.1016/s0014-4886(03)00363-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Inhibition or ablation of the hyperactive subthalamic nucleus (STN) in Parkinson's disease (PD) does not only reverse motor deficits, silencing the glutamatergic output of the subthalamic nucleus, but has also been implicated to have neuroprotective effects on nigral neurons in animal models of Parkinson's disease. Ablation of the subthalamic nucleus has been shown to increase the number of tyrosinhydroxylase-immunopositive cells and partially restores behavioral deficits in animal models of Parkinson's disease. However, it is unclear whether subthalamic nucleus ablation indeed prevents cell death or whether the effect is due to the rescue of the dopaminergic (DA) phenotype of impaired cells by upregulating tyrosine hydroxylase (TH). We therefore investigated the potential neuroprotective effects of a preceding subthalamic nucleus lesion on 6-hydroxydopamine (6-OHDA)-induced nigral cell death and compared the retrograde tracer fluorogold (FG) as a marker of cell survival with tyrosinhydroxylase immunoreactivity as a marker of the dopaminergic phenotype. In the present study, we show that ablation of the subthalamic nucleus does not affect the number of fluorogold-labeled cells but increases the number of tyrosinhydroxylase-positive neurons in subthalamic nucleus-lesioned hemiparkinsonian animals and leads to partial behavioral recovery of the rats. We conclude that subthalamic nucleus ablation exerts neuroprotective properties on the dopaminergic nigrostriatal pathway against 6-hydroxydopamine toxicity in terms of rescuing the neurotransmitter phenotype in the remaining neurons rather than enhancing the total number of nigral cells.
Collapse
Affiliation(s)
- Gesine Paul
- Department of Neurology, Charité, Humboldt-University of Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lai SK, Tse YC, Yang MS, Wong CKC, Chan YS, Yung KKL. Gene expression of glutamate receptors GluR1 and NR1 is differentially modulated in striatal neurons in rats after 6-hydroxydopamine lesion. Neurochem Int 2003; 43:639-53. [PMID: 12892651 DOI: 10.1016/s0197-0186(03)00080-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the present study, we attempted to address the modulation of the gene expression of N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate (AMPA) glutamate receptors in the neostriatum of the 6-hydroxydopamine (6-OHDA)-lesioned rat, an animal model of Parkinson's disease. After 2 weeks of lesion, reverse transcriptase-polymerase chain reactions (RT-PCRs) revealed significant reduction in GluR1 mRNA expression but a significant enhancement of NR1 mRNA expression in the striatal tissues of the lesioned side. No modulation in the mRNA expression of GluR2, GluR3, GluR4 and NR2B were found. Immunofluorescence with digital imaging analysis also demonstrated a significant reduction in GluR1 immunoreactivity in the lesioned neostriatum. Interestingly, the reduction in GluR1 immunoreactivity was primarily observed in presumed striatal medium spiny neurons but not in parvalbumin-labeled striatal GABAergic interneurons. Immunoreactivity for GluR2, GluR2/3, GluR4, NR1 and NR2B was unchanged in neurons of the neostriatum of the lesioned side. The present results indicate that there is an opposite trend in modulation in the gene expressions of GluR1 and NR1 in the neostriatum of 6-OHDA-lesioned rats after dopamine denervation. Modulation of GluR1 mRNA and immunoreactivity is likely to be limited in the striatal projection neurons. These findings have implications for the use of NMDA and AMPA receptor antagonists in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- S K Lai
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong, PR China
| | | | | | | | | | | |
Collapse
|
44
|
Guscott MR, Clarke HF, Murray F, Grimwood S, Bristow LJ, Hutson PH. The effect of (+/-)-CP-101,606, an NMDA receptor NR2B subunit selective antagonist, in the Morris watermaze. Eur J Pharmacol 2003; 476:193-9. [PMID: 12969766 DOI: 10.1016/s0014-2999(03)02182-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is well established that the NMDA receptor antagonists block hippocampal long-term potentiation and impair acquisition in the Morris watermaze task, although the role of individual NMDA receptor subtypes is largely unknown. In the present study, we compared the effects of (+/-)-CP-101,606, an antagonist selective for NMDA receptor NR1/NR2B subunit-containing receptors and the nonselective NMDA receptor antagonist MK-801, on acquisition in the Morris watermaze. Male hooded Lister rats were given 4 trials/day to find a fixed hidden platform submerged beneath the opaque water of the Morris watermaze. Twenty-four hours after the last acquisition trial, a 'probe trial' was conducted to assess the rat's spatial memory for the location of the hidden platform. Those rats treated with MK-801 (0.1 mg/kg, i.p.) 60 min prior to the acquisition and probe trials took significantly longer to find the hidden platform during training and spent significantly less time searching the platform's location during the probe trial than vehicle-treated rats. In contrast, 60-min pretreatment with (+/-)-CP-101,606 (60 mg/kg, p.o.), a dose that fully occupied hippocampal NR1/NR2B subunit-containing receptors, as determined by ex vivo NMDA receptor-specific [3H]ifenprodil binding immediately following watermaze experiments, had no effect on acquisition or the probe trial. These results suggest that antagonists selective for NR1/NR2B subunit-containing receptors may not impair spatial memory in rats in the Morris watermaze.
Collapse
Affiliation(s)
- Martin R Guscott
- Merck, Sharp and Dohme, The Neuroscience Research Centre, Behavioural Neuroscience, Terlings Park, Harlow CM20 2QR, UK.
| | | | | | | | | | | |
Collapse
|
45
|
Sousa SC, Maciel EN, Vercesi AE, Castilho RF. Ca2+-induced oxidative stress in brain mitochondria treated with the respiratory chain inhibitor rotenone. FEBS Lett 2003; 543:179-83. [PMID: 12753929 DOI: 10.1016/s0014-5793(03)00421-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this study we show that micromolar Ca(2+) concentrations (>10 microM) strongly stimulate the release of reactive oxygen species (ROS) in rotenone-treated isolated rat forebrain mitochondria. Ca(2+)-stimulated mitochondrial ROS release was associated with membrane lipid peroxidation and was directly correlated with the degree of complex I inhibition by rotenone. On the other hand, Ca(2+) did not increase mitochondrial ROS release in the presence of the complex I inhibitor 1-methyl-4-phenylpyridinium. Cyclosporin A had no effect on Ca(2+)-stimulated mitochondrial ROS release in the presence of rotenone, indicating that mitochondrial permeability transition is not involved in this process. We hypothesized that Ca(2+)-induced mitochondrial oxidative stress associated with partial inhibition of complex I may be an important factor in neuronal cell death observed in the neurodegenerative disorder Parkinson's disease.
Collapse
Affiliation(s)
- Solange C Sousa
- Departamento de Patologia Cli;nica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, SP 13083-970, Campinas, Brazil
| | | | | | | |
Collapse
|
46
|
Moro E, Esselink RJA, Benabid AL, Pollak P. Response to levodopa in parkinsonian patients with bilateral subthalamic nucleus stimulation. Brain 2002; 125:2408-17. [PMID: 12390968 DOI: 10.1093/brain/awf249] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The response to levodopa changes over time in Parkinson's disease, probably due to alterations in the dopaminergic system, progression of the disease and pulsatile oral intake of the drug. Bilateral high-frequency stimulation of the subthalamic nucleus (STN) allows a large reduction or the complete cessation of levodopa intake in patients with advanced Parkinson's disease. We studied variation in the motor short-duration response (SDR) during a levodopa challenge in bilaterally STN-stimulated patients. Twenty-eight consecutive patients with a mean duration of Parkinson's disease of 16.6 +/- 6.0 years at the time of surgery were enrolled. Fourteen patients were evaluated both before STN stimulation and 3 months after surgery (group 1) whereas the other 14 patients were assessed before implantation and after a mean of 3 years of STN stimulation (group 2). After drug withdrawal for one night, the hand-tapping test (TT) was carried out every 15 min, together with evaluation of dyskinesias using a modified Goetz scale. The Unified Parkinson's Disease Rating Scale (UPDRS) motor score was assessed every 30 min. In operated patients, STN stimulation was stopped 15 min before starting the clinical evaluations. A suprathreshold oral levodopa dose was given after one motor evaluation and two TTs. The clinical evaluation was carried out until the TT score returned to the baseline. In group 1, six patients continued without levodopa after surgery and the other eight received a daily mean dose of 337 mg; in group 2, seven patients continued without levodopa and the other seven received a daily mean dose of 386 mg. The main change in the levodopa SDR was a significant reduction in levodopa-induced dyskinesias in both groups. In those patients of group 1 who did not receive levodopa after surgery, the motor UPDRS magnitude decreased and the 'on' UPDRS motor score worsened. In group 2, the results were similar, but in the patients who continued to receive levodopa after surgery the TT magnitude increased. On the whole, chronic bilateral STN stimulation tended to decrease the magnitude of the levodopa SDR without changing the duration and latency of the response. These results suggest that continuous STN stimulation induces long-term plastic changes of the dopaminergic system, with slow and partial desensitization. In addition, the persistence of levodopa intake after surgery might hinder this beneficial process.
Collapse
Affiliation(s)
- Elena Moro
- Department of Clinical and Biological Neurosciences and INSERM U318, Joseph Fourier University, CHU de Grenoble, Grenoble, France
| | | | | | | |
Collapse
|
47
|
Stephans SE, Miller GW, Levey AI, Greenamyre JT. Acute mitochondrial and chronic toxicological effects of 1-methyl-4-phenylpyridinium in human neuroblastoma cells. Neurotoxicology 2002; 23:569-80. [PMID: 12428729 DOI: 10.1016/s0161-813x(02)00060-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
At low micromolar concentrations, 1-methyl-4-phenylpyridinium (MPP+), the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) selectively kills nigrostriatal dopaminergic neurons by mechanisms believed to involve impairment of mitochondrial complex I. A human neuroblastoma cell line expressing the dopamine transporter (DAT) was utilized to examine the effects of MPP+ on acute physiologic responses and subsequent cell death. Acute responses were measured by microphysiometry and by monitoring mitochondrial membrane potential with [3H]tetraphenylphosphonium (TPP+) uptake. MPP+ (10 microM) increased extracellular proton excretion in DAT-expressing cells within 2-3 min, but had no effect in untransfected cells. The lipophilic complex I inhibitor, rotenone, increased proton excretion in both cell lines. In DAT-expressing cells, mitochondrial membrane potential was reduced within I h of 10 microM MPP+ exposure. Rotenone reduced mitochondrial membrane potential in both cell lines. MPP+ caused apoptotic death of DAT-transfected cells 2-3 days after drug application, but did not kill untransfected cells. Thus, MPP+ produces immediate mitochondrial impairment only in cells that express DAT, and these changes occur days before overt cellular toxicity. The magnitude, time course and nature of these changes were similar to those produced by rotenone, confirming the site of action of MPP+ as mitochondrial complex I. These immediate mitochondrial effects appear to be an accurate predictor of subsequent cell death.
Collapse
Affiliation(s)
- Stacy E Stephans
- Department of Neurology, Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
48
|
Marino MJ, Awad-Granko H, Ciombor KJ, Conn PJ. Haloperidol-induced alteration in the physiological actions of group I mGlus in the subthalamic nucleus and the substantia nigra pars reticulata. Neuropharmacology 2002; 43:147-59. [PMID: 12213269 DOI: 10.1016/s0028-3908(02)00097-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Excitatory glutamatergic inputs to the subthalamic nucleus (STN), and subthalamic afferents to the substantia nigra pars reticulata (SNr) are believed to play a key role in the pathophysiology of Parkinson's disease (PD). Previously, we have shown that activation of the group I mGlus in the STN and SNr induces a direct depolarization of the neurons in these nuclei. Surprisingly, although both group I mGlus were present in the STN and SNr, mGlu5 alone mediated the DHPG-induced depolarization of the STN, and mGlu1 alone mediated the DHPG-induced depolarization of the SNr. We now report that both mGlu1 and mGlu5 are coexpressed in the same cells in both of these brain regions, and that both receptors play a role in mediating the DHPG-induced increase in intracellular calcium. Furthermore, we demonstrate that the induction of an acute PD-like state using a 16 h haloperidol treatment produces an alteration in the coupling of the group I receptors, such that post-haloperidol, DHPG-induced depolarizations are mediated by both mGlu1 and mGlu5 in the STN and SNr. Therefore, the pharmacology of the group I mGlu-mediated depolarization depends on the state of the system, and alterations in receptor coupling may be evident in pathological states such as PD.
Collapse
Affiliation(s)
- M J Marino
- Department of Neuroscience, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | |
Collapse
|
49
|
Douhou A, Debeir T, Murer MG, Do L, Dufour N, Blanchard V, Moussaoui S, Bohme GA, Agid Y, Raisman-Vozari R. Effect of chronic treatment with riluzole on the nigrostriatal dopaminergic system in weaver mutant mice. Exp Neurol 2002; 176:247-53. [PMID: 12093102 DOI: 10.1006/exnr.2002.7935] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The effects of a chronic treatment with the anti-glutamate and sodium channel modulating neuroprotective agent riluzole on the degeneration of dopamine-containing neurons were studied in the brain of weaver mutant mice. In these animals, as in Parkinson's disease, dopaminergic neurons of the nigro-striatal pathway undergo spontaneous and progressive cell death. Homozygous weaver mice were orally treated twice a day with either 8 mg/kg riluzole or placebo for 2 months. Quantification of tyrosine-hydroxylase and dopamine-transporter axonal immunostaining in the striatum revealed that riluzole significantly increased the density of striatal dopaminergic nerve terminals. These results suggest that riluzole protects dopaminergic processes in the weaver mice and/or promotes their neuroplasticity.
Collapse
Affiliation(s)
- Aïcha Douhou
- INSERM U-289, Hôpital de la Salpétriêre, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Parkinson's disease (PD) is a common and disabling neurodegenerative disease marked by progressive motor dysfunction, which results from selective degeneration of the nigrostriatal pathway. Epidemiological studies indicate that exposure to pesticides, rural living, farming, and drinking well water are associated with an increased risk of developing PD. Rare cases of PD are caused by mutations in nuclear genes, and there is increasing evidence for susceptibility genes that alter disease risk. Parkinson's disease is also associated with a systemic defect in mitochondrial complex I activity. Animal models indicate that exposure to inhibitors of mitochondrial complex I, including pesticides, is sufficient to reproduce the features of PD, but genetic factors clearly modulate susceptibility. Complex I defects may result in oxidative stress and increase the susceptibility of neurons to excitotoxic death. In this way, environmental exposures and mitochondrial dysfunction may interact and result in neurodegeneration.
Collapse
Affiliation(s)
- Todd B Sherer
- Department of Neurology, Emory University, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|