1
|
Kang W, Frouni I, Bédard D, Kwan C, Hamadjida A, Nuara SG, Gourdon JC, Huot P. Positive allosteric mGluR 2 modulation with BINA alleviates dyskinesia and psychosis-like behaviours in the MPTP-lesioned marmoset. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8917-8924. [PMID: 38861009 DOI: 10.1007/s00210-024-03215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
There is mounting evidence that positive allosteric modulation of metabotropic glutamate type 2 receptors (mGluR2) is an efficacious approach to reduce the severity of L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia, psychosis-like behaviours (PLBs), while conferring additional anti-parkinsonian benefit. However, the mGluR2 positive allosteric modulators (PAMs) tested so far, LY-487,379 and CBiPES, share a similar chemical scaffold. Here, we sought to assess whether similar benefits would be conferred by a structurally-distinct mGluR2 PAM, biphenylindanone A (BINA). Six 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned marmosets exhibiting dyskinesia and PLBs were administered L-DOPA with either vehicle or BINA (0.1, 1, and 10 mg/kg) in a randomised within-subject design and recorded. Behaviour was analysed by a blinded rater who scored the severity of each of parkinsonism, dyskinesia and PLBs. When added to L-DOPA, BINA 0.1 mg/kg, 1 mg/kg, and 10 mg/kg all significantly reduced the severity of global dyskinesia, by 40%, 52% and 53%, (all P < 0.001) respectively. BINA similarly attenuated the severity of global PLBs by 35%, 48%, and 50%, (all P < 0.001) respectively. Meanwhile, BINA did not alter the effect of L-DOPA on parkinsonism exhibited by the marmosets. The results of this study provide incremental evidence of positive allosteric modulation of mGluR2 as an effective therapeutic strategy for alleviating dyskinesia and PLBs, without hindering the anti-parkinsonian action of L-DOPA. Furthermore, this therapeutic benefit does not appear to be confined to a particular chemical scaffold.
Collapse
Affiliation(s)
- Woojin Kang
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Dominique Bédard
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Cynthia Kwan
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Adjia Hamadjida
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Stephen G Nuara
- Comparative Medicine & Animal Resource Centre, McGill University, Montreal, QC, Canada
| | - Jim C Gourdon
- Comparative Medicine & Animal Resource Centre, McGill University, Montreal, QC, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada.
- Comparative Medicine & Animal Resource Centre, McGill University, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Department of Neurosciences, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
2
|
Doppler CEJ, Seger A, Farrher E, Régio Brambilla C, Hensel L, Filss CP, Hellmich M, Gogishvili A, Shah NJ, Lerche CW, Neumaier B, Langen KJ, Fink GR, Sommerauer M. Glutamate Signaling in Patients With Parkinson Disease With REM Sleep Behavior Disorder. Neurology 2024; 102:e209271. [PMID: 38630966 DOI: 10.1212/wnl.0000000000209271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Clinical heterogeneity of patients with Parkinson disease (PD) is well recognized. PD with REM sleep behavior disorder (RBD) is a more malignant phenotype with faster motor progression and higher nonmotor symptom burden. However, the neural mechanisms underlying this clinical divergence concerning imbalances in neurotransmitter systems remain elusive. METHODS Combining magnetic resonance (MR) spectroscopy and [11C]ABP688 PET on a PET/MR hybrid system, we simultaneously investigated two different mechanisms of glutamate signaling in patients with PD. Patients were grouped according to their RBD status in overnight video-polysomnography and compared with age-matched and sex-matched healthy control (HC) participants. Total volumes of distribution (VT) of [11C]ABP688 were estimated with metabolite-corrected plasma concentrations during steady-state conditions between 45 and 60 minutes of the scan following a bolus-infusion protocol. Glutamate, glutamine, and glutathione levels were investigated with single-voxel stimulated echo acquisition mode MR spectroscopy of the left basal ganglia. RESULTS We measured globally elevated VT of [11C]ABP688 in 16 patients with PD and RBD compared with 17 patients without RBD and 15 HC participants (F(2,45) = 5.579, p = 0.007). Conversely, glutamatergic metabolites did not differ between groups and did not correlate with the regional VT of [11C]ABP688. VT of [11C]ABP688 correlated with the amount of REM sleep without atonia (F(1,42) = 5.600, p = 0.023) and with dopaminergic treatment response in patients with PD (F(1,30) = 5.823, p = 0.022). DISCUSSION Our results suggest that patients with PD and RBD exhibit altered glutamatergic signaling indicated by higher VT of [11C]ABP688 despite unaffected glutamate levels. The imbalance of glutamate receptors and MR spectroscopy glutamate metabolite levels indicates a novel mechanism contributing to the heterogeneity of PD and warrants further investigation of drugs targeting mGluR5.
Collapse
Affiliation(s)
- Christopher E J Doppler
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Aline Seger
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ezequiel Farrher
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Cláudia Régio Brambilla
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Lukas Hensel
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christian P Filss
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Martin Hellmich
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ana Gogishvili
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - N Jon Shah
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christoph W Lerche
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Bernd Neumaier
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Karl-Josef Langen
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Gereon R Fink
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Michael Sommerauer
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| |
Collapse
|
3
|
Malyshev AV, Pavshintcev VV, Mitkin NA, Sukhanova IA, Gedzun VR, Zlobin AS, Doronin II, Babkin GA, Sawyer TK. The novel peptide LCGM-10 attenuates metabotropic glutamate receptor 5 activity and demonstrates behavioral effects in animal models. Front Behav Neurosci 2024; 18:1333258. [PMID: 38385004 PMCID: PMC10879279 DOI: 10.3389/fnbeh.2024.1333258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
We employed a structural bioinformatics approach to develop novel peptides with predicted affinity to the binding site for negative allosteric modulators (NAMs) of metabotropic glutamate receptor 5 (mGluR5). Primary screening in zebrafish (Danio rerio) revealed a stimulatory effect of two peptides, LCGM-10 and LCGM-15. Target validation studies using calcium ion flux imaging and a luciferase reporter assay confirmed mGluR5 as the target. LCGM-10 showed greater potency than LCGM-15; it was comparable to that of the mGluR5 NAM 2-methyl-6-(phenylethynyl) pyridine (MPEP). Rodent behavioral screening in the open field and elevated plus maze revealed increased locomotor activity in both tests after acute LCGM-10 treatment, supported by further analysis of home cage spontaneous locomotor activity (SLA). The stimulating effect of a single LCGM-10 administration on SLA was evident up to 60 min after administration and was not accompanied by hypokinetic rebound observed for caffeine. According to our results, LCGM-10 has therapeutic potential to treat hypo- and dyskinesias of various etiologies. Further investigation of LCGM-10 effects in the delay discounting model of impulsive choice in rats revealed reduced trait impulsivity after single and chronic administrations, suggesting potential implication for attention deficit hyperactivity disorder, obsessive compulsive disorder, and addictions.
Collapse
|
4
|
Kochoian BA, Bure C, Papa SM. Targeting Striatal Glutamate and Phosphodiesterases to Control L-DOPA-Induced Dyskinesia. Cells 2023; 12:2754. [PMID: 38067182 PMCID: PMC10706484 DOI: 10.3390/cells12232754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
A large body of work during the past several decades has been focused on therapeutic strategies to control L-DOPA-induced dyskinesias (LIDs), common motor complications of long-term L-DOPA therapy in Parkinson's disease (PD). Yet, LIDs remain a clinical challenge for the management of patients with advanced disease. Glutamatergic dysregulation of striatal projection neurons (SPNs) appears to be a key contributor to altered motor responses to L-DOPA. Targeting striatal hyperactivity at the glutamatergic neurotransmission level led to significant preclinical and clinical trials of a variety of antiglutamatergic agents. In fact, the only FDA-approved treatment for LIDs is amantadine, a drug with NMDAR antagonistic actions. Still, novel agents with improved pharmacological profiles are needed for LID therapy. Recently other therapeutic targets to reduce dysregulated SPN activity at the signal transduction level have emerged. In particular, mechanisms regulating the levels of cyclic nucleotides play a major role in the transduction of dopamine signals in SPNs. The phosphodiesterases (PDEs), a large family of enzymes that degrade cyclic nucleotides in a specific manner, are of special interest. We will review the research for antiglutamatergic and PDE inhibition strategies in view of the future development of novel LID therapies.
Collapse
Affiliation(s)
- Brik A. Kochoian
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Cassandra Bure
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Stella M. Papa
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30329, USA
| |
Collapse
|
5
|
Mango D, Ledonne A. Updates on the Physiopathology of Group I Metabotropic Glutamate Receptors (mGluRI)-Dependent Long-Term Depression. Cells 2023; 12:1588. [PMID: 37371058 DOI: 10.3390/cells12121588] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Group I metabotropic glutamate receptors (mGluRI), including mGluR1 and mGluR5 subtypes, modulate essential brain functions by affecting neuronal excitability, intracellular calcium dynamics, protein synthesis, dendritic spine formation, and synaptic transmission and plasticity. Nowadays, it is well appreciated that the mGluRI-dependent long-term depression (LTD) of glutamatergic synaptic transmission (mGluRI-LTD) is a key mechanism by which mGluRI shapes connectivity in various cerebral circuitries, directing complex brain functions and behaviors, and that it is deranged in several neurological and psychiatric illnesses, including neurodevelopmental disorders, neurodegenerative diseases, and psychopathologies. Here, we will provide an updated overview of the physiopathology of mGluRI-LTD, by describing mechanisms of induction and regulation by endogenous mGluRI interactors, as well as functional physiological implications and pathological deviations.
Collapse
Affiliation(s)
- Dalila Mango
- School of Pharmacy, University of Rome "Tor Vergata", 00133 Rome, Italy
- Laboratory of Pharmacology of Synaptic Plasticity, European Brain Research Institute, 00161 Rome, Italy
| | - Ada Ledonne
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
6
|
Epping-Jordan MP, Girard F, Bessis AS, Mutel V, Boléa C, Derouet F, Bessif A, Mingard B, Barbier S, Paradis JS, Rocher JP, Lütjens R, Kalinichev M, Poli S. Effect of the Metabotropic Glutamate Receptor Type 5 Negative Allosteric Modulator Dipraglurant on Motor and Non-Motor Symptoms of Parkinson's Disease. Cells 2023; 12:1004. [PMID: 37048075 PMCID: PMC10093229 DOI: 10.3390/cells12071004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson's disease (PD) patients suffer not only from the primary motor symptoms of the disease but also from a range of non-motor symptoms (NMS) that cause disability and low quality of life. Excessive glutamate activity in the basal ganglia resulting from degeneration of the nigrostriatal dopamine pathway has been implicated in the motor symptoms, NMS and dyskinesias in PD patients. In this study, we investigated the effects of a selective mGlu5 negative allosteric modulator (NAM), dipraglurant, in a rodent motor symptoms model of PD, but also in models of anxiety, depression and obsessive-compulsive disorder, all of which are among the most prevalent NMS symptoms. Dipraglurant is rapidly absorbed after oral administration, readily crosses the blood-brain barrier, and exhibits a high correlation between plasma concentration and efficacy in behavioral models. In vivo, dipraglurant dose-dependently reduced haloperidol-induced catalepsy, increased punished licks in the Vogel conflict-drinking model, decreased immobility time in the forced swim test, decreased the number of buried marbles in the marble-burying test, but had no effect on rotarod performance or locomotor activity. These findings suggest that dipraglurant may have benefits to address some of the highly problematic comorbid non-motor symptoms of PD, in addition to its antidyskinetic effect demonstrated in PD-LID patients.
Collapse
|
7
|
Kwan C, Kang W, Kim E, Belliveau S, Frouni I, Huot P. Metabotropic glutamate receptors in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:1-31. [PMID: 36868628 DOI: 10.1016/bs.irn.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Parkinson's disease (PD) is a complex disorder that leads to alterations in multiple neurotransmitter systems, notably glutamate. As such, several drugs acting at glutamatergic receptors have been assessed to alleviate the manifestation of PD and treatment-related complications, culminating with the approval of the N-methyl-d-aspartate (NMDA) antagonist amantadine for l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia. Glutamate elicits its actions through several ionotropic and metabotropic (mGlu) receptors. There are 8 sub-types of mGlu receptors, with sub-types 4 (mGlu4) and 5 (mGlu5) modulators having been tested in the clinic for endpoints pertaining to PD, while sub-types 2 (mGlu2) and 3 (mGlu3) have been investigated in pre-clinical settings. In this book chapter, we provide an overview of mGlu receptors in PD, with a focus on mGlu5, mGlu4, mGlu2 and mGlu3 receptors. For each sub-type, we review, when applicable, their anatomical localization and possible mechanisms underlying their efficacy for specific disease manifestation or treatment-induced complications. We then summarize the findings of pre-clinical studies and clinical trials with pharmacological agents and discuss the potential strengths and limitations of each target. We conclude by offering some perspectives on the potential use of mGlu modulators in the treatment of PD.
Collapse
Affiliation(s)
- Cynthia Kwan
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Woojin Kang
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Esther Kim
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Sébastien Belliveau
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Movement Disorder Clinic, Division of Neurology, Department of Neurosciences, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
8
|
Targeting G Protein-Coupled Receptors in the Treatment of Parkinson's Disease. J Mol Biol 2022:167927. [PMID: 36563742 DOI: 10.1016/j.jmb.2022.167927] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized in part by the deterioration of dopaminergic neurons which leads to motor impairment. Although there is no cure for PD, the motor symptoms can be treated using dopamine replacement therapies including the dopamine precursor L-DOPA, which has been in use since the 1960s. However, neurodegeneration in PD is not limited to dopaminergic neurons, and many patients experience non-motor symptoms including cognitive impairment or neuropsychiatric disturbances, for which there are limited treatment options. Moreover, there are currently no treatments able to alter the progression of neurodegeneration. There are many therapeutic strategies being investigated for PD, including alternatives to L-DOPA for the treatment of motor impairment, symptomatic treatments for non-motor symptoms, and neuroprotective or disease-modifying agents. G protein-coupled receptors (GPCRs), which include the dopamine receptors, are highly druggable cell surface proteins which can regulate numerous intracellular signaling pathways and thereby modulate the function of neuronal circuits affected by PD. This review will describe the treatment strategies being investigated for PD that target GPCRs and their downstream signaling mechanisms. First, we discuss new developments in dopaminergic agents for alleviating PD motor impairment, the role of dopamine receptors in L-DOPA induced dyskinesia, as well as agents targeting non-dopamine GPCRs which could augment or replace traditional dopaminergic treatments. We then discuss GPCRs as prospective treatments for neuropsychiatric and cognitive symptoms in PD. Finally, we discuss the evidence pertaining to ghrelin receptors, β-adrenergic receptors, angiotensin receptors and glucagon-like peptide 1 receptors, which have been proposed as disease modifying targets with potential neuroprotective effects in PD.
Collapse
|
9
|
Frouni I, Huot P. Glutamate modulation for the treatment of levodopa induced dyskinesia: a brief review of the drugs tested in the clinic. Neurodegener Dis Manag 2022; 12:203-214. [PMID: 35587024 DOI: 10.2217/nmt-2021-0055] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Levodopa is the standard treatment for Parkinson's disease, but its use is marred by the emergence of dyskinesia, for which treatment options remain limited. Here, we review the glutamatergic modulators that were assessed for their antidyskinetic potential in clinical trials, including N-methyl-D-aspartate (NMDA) antagonists, agonists at the glycine-binding site on NMDA receptors, metabotropic glutamate (mGlu) 4 agonists, mGlu5 antagonists, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonists and glutamate release inhibitors. Several agents that were investigated are not selective for their targets, raising uncertainty about the extent to which glutamatergic modulation contributed to their effects. Except for amantadine, the use of glutamatergic modulators for the treatment of dyskinesia in Parkinson's disease remains largely investigational, with promising results obtained with mGlu5 negative allosteric modulation.
Collapse
Affiliation(s)
- Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada.,Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Neuroscience, Division of Neurology, McGill University Health Centre, Montreal, QC, H3A 2B4, Canada
| |
Collapse
|
10
|
Azam S, Jakaria M, Kim J, Ahn J, Kim IS, Choi DK. Group I mGluRs in Therapy and Diagnosis of Parkinson’s Disease: Focus on mGluR5 Subtype. Biomedicines 2022; 10:biomedicines10040864. [PMID: 35453614 PMCID: PMC9032558 DOI: 10.3390/biomedicines10040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs; members of class C G-protein-coupled receptors) have been shown to modulate excitatory neurotransmission, regulate presynaptic extracellular glutamate levels, and modulate postsynaptic ion channels on dendritic spines. mGluRs were found to activate myriad signalling pathways to regulate synapse formation, long-term potentiation, autophagy, apoptosis, necroptosis, and pro-inflammatory cytokines release. A notorious expression pattern of mGluRs has been evident in several neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and schizophrenia. Among the several mGluRs, mGluR5 is one of the most investigated types of considered prospective therapeutic targets and potential diagnostic tools in neurodegenerative diseases and neuropsychiatric disorders. Recent research showed mGluR5 radioligands could be a potential tool to assess neurodegenerative disease progression and trace respective drugs’ kinetic properties. This article provides insight into the group I mGluRs, specifically mGluR5, in the progression and possible therapy for PD.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Md. Jakaria
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - JoonSoo Kim
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Jaeyong Ahn
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
11
|
Angela Cenci M, Skovgård K, Odin P. Non-dopaminergic approaches to the treatment of motor complications in Parkinson's disease. Neuropharmacology 2022; 210:109027. [DOI: 10.1016/j.neuropharm.2022.109027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/21/2022]
|
12
|
Gu L, Luo WY, Xia N, Zhang JN, Fan JK, Yang HM, Wang MC, Zhang H. Upregulated mGluR5 induces ER stress and DNA damage by regulating the NMDA receptor subunit NR2B. J Biochem 2021; 171:349-359. [PMID: 34908130 DOI: 10.1093/jb/mvab140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/06/2021] [Indexed: 11/12/2022] Open
Abstract
Dysfunction caused by mGluR5 expression or activation is an important mechanism in the development of Parkinson's disease (PD). Early clinical studies on mGluR5 negative allosteric modulators have shown some limitations. It is therefore necessary to find a more specific approach to block mGluR5-mediated neurotoxicity. Here, we determined the role of NMDA receptor subunit NR2B in mGluR5-mediated ER stress and DNA damage. In vitro study, rotenone-induced ER stress and DNA damage were accompanied by an increase in mGluR5 expression, and overexpressed or activated mGluR5 with agonist CHPG induced ER stress and DNA damage, while blocking mGluR5 with antagonist MPEP alleviated the effect. Furthermore, the damage caused by CHPG was blocked by NMDA receptor antagonist MK-801. Additionally, rotenone or CHPG increased the p-Src and p-NR2B, which was inhibited by MPEP. Blocking p-Src or NR2B with PP2 or CP101,606 alleviated CHPG-induced ER stress and DNA damage. Overactivation of mGluR5 accompanied with the increase of p-Src and p-NR2B in the ER stress and DNA damage was found in rotenone-induced PD rat model. These findings suggest a new mechanism wherein mGluR5 induces ER stress and DNA damage through the NMDA receptor and propose NR2B as the molecular target for therapeutic strategy for PD.
Collapse
Affiliation(s)
- Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Wen-Yuan Luo
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ning Xia
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.,Department of neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jing-Kai Fan
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Meng-Chen Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
13
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Rescue of striatal long-term depression by chronic mGlu5 receptor negative allosteric modulation in distinct dystonia models. Neuropharmacology 2021; 192:108608. [PMID: 33991565 DOI: 10.1016/j.neuropharm.2021.108608] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
An impairment of long-term synaptic plasticity is considered as a peculiar endophenotype of distinct forms of dystonia, a common, disabling movement disorder. Among the few therapeutic options, broad-spectrum antimuscarinic drugs are utilized, aimed at counteracting abnormal striatal acetylcholine-mediated transmission, which plays a crucial role in dystonia pathophysiology. We previously demonstrated a complete loss of long-term synaptic depression (LTD) at corticostriatal synapses in rodent models of two distinct forms of isolated dystonia, resulting from mutations in the TOR1A (DYT1), and GNAL (DYT25) genes. In addition to anticholinergic agents, the aberrant excitability of striatal cholinergic cells can be modulated by group I metabotropic glutamate receptor subtypes (mGlu1 and 5). Here, we tested the efficacy of the negative allosteric modulator (NAM) of metabotropic glutamate 5 (mGlu) receptor, dipraglurant (ADX48621) on striatal LTD. We show that, whereas acute treatment failed to rescue LTD, chronic dipraglurant rescued this form of synaptic plasticity both in DYT1 mice and GNAL rats. Our analysis of the pharmacokinetic profile of dipraglurant revealed a relatively short half-life, which led us to uncover a peculiar time-course of recovery based on the timing from last dipraglurant injection. Indeed, striatal spiny projection neurons (SPNs) recorded within 2 h from last administration showed full expression of synaptic plasticity, whilst the extent of recovery progressively diminished when SPNs were recorded 4-6 h after treatment. Our findings suggest that distinct dystonia genes may share common signaling pathway dysfunction. More importantly, they indicate that dipraglurant might be a potential novel therapeutic agent for this disabling disorder.
Collapse
|
15
|
Chen X, Wang Y, Wu H, Cheng C, Le W. Research advances on L-DOPA-induced dyskinesia: from animal models to human disease. Neurol Sci 2020; 41:2055-2065. [DOI: 10.1007/s10072-020-04333-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/07/2020] [Indexed: 02/06/2023]
|
16
|
Roles of Glutamate Receptors in Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20184391. [PMID: 31500132 PMCID: PMC6769661 DOI: 10.3390/ijms20184391] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder resulting from the degeneration of pigmented dopaminergic neurons in the substantia nigra pars compacta. It induces a series of functional modifications in the circuitry of the basal ganglia nuclei and leads to severe motor disturbances. The amino acid glutamate, as an excitatory neurotransmitter, plays a key role in the disruption of normal basal ganglia function regulated through the interaction with its receptor proteins. It has been proven that glutamate receptors participate in the modulation of neuronal excitability, transmitter release, and long-term synaptic plasticity, in addition to being related to the altered neurotransmission in Parkinson's disease. Therefore, they are considered new targets for improving the therapeutic strategies used to treat Parkinson's disease. In this review, we discuss the biological characteristics of these receptors and demonstrate the receptor-mediated neuroprotection in Parkinson's disease. Pharmacological manipulation of these receptors during anti-Parkinsonian processes in both experimental studies and clinical trials are also summarized.
Collapse
|
17
|
Huang Y, Shu H, Li L, Zhen T, Zhao J, Zhou X, Luo W. L-DOPA-Induced Motor Impairment and Overexpression of Corticostriatal Synaptic Components Are Improved by the mGluR5 Antagonist MPEP in 6-OHDA-Lesioned Rats. ASN Neuro 2019; 10:1759091418811021. [PMID: 30439288 PMCID: PMC6238196 DOI: 10.1177/1759091418811021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Levodopa (L-DOPA) is still the most effective drug for the treatment of Parkinson's disease (PD). However, the long-term therapy often triggers L-DOPA-induced dyskinesia (LID). Metabotropic glutamate receptor type 5 (mGluR5) is abundant in the basal ganglia, and its inhibition is thought to modulate postsynaptic excitatory synaptic transmission and glutamate hyperactivity in PD and LID. In this report, we examined the effects of mGluR5-specific antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) on LID and synaptic components in the PD model rat. We found the selective mGluR5 antagonist MPEP attenuated abnormal involuntary movements, prolonged the duration of rotational response, reversed the decrease of left forepaw adjusting steps, and reduced overexpression of striatal mGluR5 in the LID rats. Moreover, our results showed much thicker postsynaptic densities, narrower synapse cleft, as well as the increased ratio of perforated synapses induced by L-DOPA treatment, while coadministration of L-DOPA and MPEP reversed these postsynaptic effects. Finally, MPEP reduced overexpression of the two postsynaptic proteins (PSD-95 and SAP102) induced by L-DOPA treatment. Hence, these results provide evidence that aberrant neural plasticity at corticostriatal synapses in the striatum is closely correlated with the occurrence of LID, and targeted inhibition of mGluR5 by MPEP alleviates LID in the PD rat model.
Collapse
Affiliation(s)
- Yixian Huang
- 1 Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiyang Shu
- 2 Department of Neurology, Taikang People's Hospital of Henan Province, Taikang, China
| | - Li Li
- 1 Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tili Zhen
- 1 Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junyan Zhao
- 1 Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianju Zhou
- 4 Laboratory of Neurological Diseases, Department of Neurology, Changzhou No.2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Weifeng Luo
- 1 Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,3 Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
18
|
Luo WY, Xing SQ, Zhu P, Zhang CG, Yang HM, Van Halm-Lutterodt N, Gu L, Zhang H. PDZ Scaffold Protein CAL Couples with Metabotropic Glutamate Receptor 5 to Protect Against Cell Apoptosis and Is a Potential Target in the Treatment of Parkinson's Disease. Neurotherapeutics 2019; 16:761-783. [PMID: 31073978 PMCID: PMC6694344 DOI: 10.1007/s13311-019-00730-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Targeting mGluR5 has been an attractive strategy to modulate glutamate excitotoxicity for neuroprotection. Although human clinical trials using mGluR5 negative allosteric modulators (NAMs) have included some disappointments, recent investigations have added several more attractive small molecules to this field, providing a promise that the identification of more additional strategies to modulate mGluR5 activity might be potentially beneficial for the advancement of PD treatment. Here, we determined the role of the interacting partner CAL (cystic fibrosis transmembrane conductance regulator-associated ligand) in mGluR5-mediated protection in vitro and in vivo. In astroglial C6 cells, CAL deficiency blocked (S)-3, 5-dihydroxyphenylglycine (DHPG)-elicited p-AKT and p-ERK1/2, subsequently prevented group I mGluRs-mediated anti-apoptotic protection, which was blocked by receptor antagonist 1-aminoindan-1, 5-dicarboxylic acid (AIDA), and PI3K or MEK inhibitor LY294002 or U0126. In rotenone-treated MN9D cells, both CAL and mGluR5 expressions were decreased in a time- and dose-dependent manner, and the correlation between these 2 proteins was confirmed by lentivirus-delivered CAL overexpression and knockdown. Moreover, CAL coupled with mGluR5 upregulated mGluR5 protein expression by inhibition of ubiquitin-proteasome-dependent degradation to suppress mGluR5-mediated p-JNK and to protect against cell apoptosis. Additionally, CAL also inhibited rotenone-induced glutamate release to modulate mGluR5 activity. Furthermore, in the rotenone-induced rat model of PD, AAV-delivered CAL overexpression attenuated behavioral deficits and dopaminergic neuronal death, while CAL deficiency aggravated rotenone toxicity. On the other hand, the protective effect of the mGluR5 antagonist MPEP was weakened by knocking down CAL. In vivo experiments also confirmed that CAL inhibited ubiquitination-proteasome-dependent degradation to modulate mGluR5 expression and JNK phosphorylation. Our findings show that CAL protects against cell apoptosis via modulating mGluR5 activity, and may be a new molecular target for an effective therapeutic strategy for PD.
Collapse
Affiliation(s)
- Wen Yuan Luo
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Su Qian Xing
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ping Zhu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Chen Guang Zhang
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, 100069, China
| | - Hui Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Nicholas Van Halm-Lutterodt
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100069, China
- Department of Orthopaedics and Neurosurgery, Keck Medical Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China.
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
19
|
Pourmirbabaei S, Dolatshahi M, Rahmani F. Pathophysiological clues to therapeutic applications of glutamate mGlu5 receptor antagonists in levodopa-induced dyskinesia. Eur J Pharmacol 2019; 855:149-159. [PMID: 31063776 DOI: 10.1016/j.ejphar.2019.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/16/2022]
Abstract
Levodopa remains to be the mainstay for treatment of Parkinson disease (PD). Long-term levodopa treatment bears a risk for developing levodopa-induced dyskinesia (LID). LID significantly overshadows patients' quality of life and therapeutic efficacy of levodopa. Pre- and post-synaptic changes in dopamine secretion and signaling, along with altered glutamate receptor expression and glutamatergic signaling in striatal neurons, and the resulting disinhibition-like changes in the corticostriatal circuitry, lead to aberrant activity of motor cortex and formation of LID. Research has highlighted the role of group I metabotropic glutamate receptors especially the metabotropic glutamate receptor 5 (mGlu5) in formation of LID through potentiating of ionotropic glutamate NMDA receptors and dopamine D1/D5 receptors in direct pathway. Accordingly, MTEP and MPEP were the first mGlu5 receptor antagonists which were shown to attenuate LID in animal models through suppression of downstream signaling cascades involving mitogen-activated protein kinase (MAPK) and FosB/delta FosB activation, as well as modulation of prodynorphinegic, preproenkephalinergic, and GABA-ergic neurotransmission systems. Beneficial effects of other mGlu5 receptor antagonists such as AFQ056/mavoglurant and ADX48621/dipraglurant in amelioration of LID has been shown not only in animal models but also in clinical trials. Considering the presence of mGlu receptor dysregulation in rapid eye movement (REM) sleep behavior disorder and depression, which are prodromal signs of PD, along with the neuroprotective effects of mGlu receptor antagonists, and their cognitive benefits, potential effectiveness of mGlu receptor antagonists in early prevention of PD remains to be investigated.
Collapse
Affiliation(s)
- Shayan Pourmirbabaei
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Dolatshahi
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Rahmani
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
20
|
Charvin D, Medori R, Hauser RA, Rascol O. Therapeutic strategies for Parkinson disease: beyond dopaminergic drugs. Nat Rev Drug Discov 2018; 17:804-822. [PMID: 30262889 DOI: 10.1038/nrd.2018.136] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Existing therapeutic strategies for managing Parkinson disease (PD), which focus on addressing the loss of dopamine and dopaminergic function linked with degeneration of dopaminergic neurons, are limited by side effects and lack of long-term efficacy. In recent decades, research into PD pathophysiology and pharmacology has focused on understanding and tackling the neurodegenerative processes and symptomology of PD. In this Review, we discuss the challenges associated with the development of novel therapies for PD, highlighting emerging agents that aim to target cell death, as well as new targets offering a symptomatic approach to managing features and progression of the disease.
Collapse
Affiliation(s)
| | | | - Robert A Hauser
- Department of Neurology, University of South Florida, Tampa, FL, USA
| | - Olivier Rascol
- Centre d'Investigation Clinique CIC1436, Services de Neurologie et de Pharmacologie Clinique, Réseau NS-PARK/FCRIN et Centre COEN NeuroToul, CHU de Toulouse, INSERM, University of Toulouse 3, Toulouse, France
| |
Collapse
|
21
|
Genetic Knockdown of mGluR5 in Striatal D1R-Containing Neurons Attenuates L-DOPA-Induced Dyskinesia in Aphakia Mice. Mol Neurobiol 2018; 56:4037-4050. [PMID: 30259400 DOI: 10.1007/s12035-018-1356-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
L-DOPA is the main pharmacological therapy for Parkinson's disease. However, long-term exposure to L-DOPA induces involuntary movements termed dyskinesia. Clinical trials show that dyskinesia is attenuated by metabotropic glutamate receptor type 5 (mGluR5) antagonists. Further, the onset of dyskinesia is delayed by nicotine and mGluR5 expression is lower in smokers than in non-smokers. However, the mechanisms by which mGluR5 modulates dyskinesia and how mGluR5 and nicotine interact have not been established. To address these issues, we studied the role of mGluR5 in D1R-containing neurons in dyskinesia and examined whether nicotine reduces dyskinesia via mGluR5. In the aphakia mouse model of Parkinson's disease, we selectively knocked down mGluR5 in D1R-containing neurons (aphakia-mGluR5KD-D1). We found that genetic downregulation of mGluR5 decreased dyskinesia in aphakia mice. Although chronic nicotine increased the therapeutic effect of L-DOPA in both aphakia and aphakia-mGluR5KD-D1 mice, it caused a robust reduction in dyskinesia only in aphakia, and not in aphakia-mGluR5KD-D1 mice. Downregulating mGluR5 or nicotine treatment after L-DOPA decreased ERK and histone 3 activation, and FosB expression. Combining nicotine and mGluR5 knockdown did not have an added antidyskinetic effect, indicating that the effect of nicotine might be mediated by downregulation of mGluR5 expression. Treatment of aphakia-mGluR5KD-D1 mice with a negative allosteric modulator did not further modify dyskinesia, suggesting that mGluR5 in non-D1R-containing neurons does not play a role in its development. In conclusion, this work suggests that mGluR5 antagonists reduce dyskinesia by mainly affecting D1R-containing neurons and that the effect of nicotine on dyskinetic signs in aphakia mice is likely via mGluR5.
Collapse
|
22
|
Charvin D, Di Paolo T, Bezard E, Gregoire L, Takano A, Duvey G, Pioli E, Halldin C, Medori R, Conquet F. An mGlu4-Positive Allosteric Modulator Alleviates Parkinsonism in Primates. Mov Disord 2018; 33:1619-1631. [PMID: 30216534 DOI: 10.1002/mds.27462] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 04/19/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Levodopa remains the gold-standard treatment for PD. However, it becomes less effective as the disease progresses and produces debilitating side effects, such as motor fluctuations and l-dopa-induced dyskinesia. Modulation of metabotropic glutamate receptor 4 represents a promising antiparkinsonian approach in combination with l-dopa, but it has not been demonstrated in primates. OBJECTIVE We studied whether a novel positive allosteric modulator of the metabotropic glutamate receptor 4, PXT002331 (foliglurax), could reduce parkinsonism in primate models. METHODS We assessed the therapeutic potential of PXT002331 in three models of MPTP-induced parkinsonism in macaques. These models represent three different stages of disease evolution: early stage and advanced stage with and without l-dopa-induced dyskinesia. RESULTS As an adjunct to l-dopa, PXT002331 induced a robust and dose-dependent reversal of parkinsonian motor symptoms in macaques, including bradykinesia, tremor, posture, and mobility. Moreover, PXT002331 strongly decreased dyskinesia severity, thus having therapeutic efficacy on both parkinsonian motor impairment and l-dopa-induced dyskinesia. PXT002331 brain penetration was also assessed using PET imaging in macaques, and pharmacodynamic analyses support target engagement in the therapeutic effects of PXT002331. CONCLUSIONS This work provides a demonstration that a positive allosteric modulator of metabotropic glutamate receptor 4 can alleviate the motor symptoms of PD and the motor complications induced by l-dopa in primates. PXT002331 is the first compound of its class to enter phase IIa clinical trials. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Delphine Charvin
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Therese Di Paolo
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Erwan Bezard
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Laurent Gregoire
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Akihiro Takano
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Guillaume Duvey
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Elsa Pioli
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Christer Halldin
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Rossella Medori
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - François Conquet
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| |
Collapse
|
23
|
Veyres N, Hamadjida A, Huot P. Predictive Value of Parkinsonian Primates in Pharmacologic Studies: A Comparison between the Macaque, Marmoset, and Squirrel Monkey. J Pharmacol Exp Ther 2018. [DOI: 10.1124/jpet.117.247171] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
24
|
Gupta I, Young AMJ. Metabotropic glutamate receptor modulation of dopamine release in the nucleus accumbens shell is unaffected by phencyclidine pretreatment: In vitro assessment using fast-scan cyclic voltammetry rat brain slices. Brain Res 2018. [PMID: 29524437 DOI: 10.1016/j.brainres.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The non-competitive glutamate antagonist, phencyclidine is used in rodents to model behavioural deficits see in schizophrenia. Importantly, these deficits endure long after the cessation of short-term chronic treatment (sub-chronic), indicating that the drug treatment causes long-term changes in the physiology and/or chemistry of the brain. There is evidence that this may occur through glutamatergic modulation of mesolimbic dopamine release, perhaps involving metabotropic glutamate receptors (mGluR). This study sought to investigate the effect of sub-chronic phencyclidine pretreatment on modulation of dopamine neurotransmission by metabotropic glutamate receptors 2 and 5 (mGluR2 and mGluR5) in the nucleus accumbens shell in vitro, with the hypothesis that phencyclidine pretreatment would disrupt the mGluR-mediated modulation of dopamine release. We showed that the orthosteric mGluR2 agonist LY379268 (0.1 µM, 1 µM and 10 µM) and mGluR5 positive allosteric modulator CDPPB (1 µM and 10 µM) both attenuated potassium-evoked dopamine release, underscoring their role in modulating dopamine neurotransmission in the nucleus accumbens. Sub-chronic PCP treatment, which caused cognitive deficits measured by performance in the novel object recognition task, modelling aspects of behavioral deficits seen in schizophrenia, induced neurobiological changes that enhanced dopamine release in the nucleus accumbens, but had no effect on mGluR2 or mGluR5 mediated changes in dopamine release. Therefore it is unlikely that schizophrenia-related behavioural changes seen after sub-chronic phencyclidine pre-treatment are mediated through mGluR modulation of dopamine release.
Collapse
Affiliation(s)
- Ishan Gupta
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Andrew M J Young
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK.
| |
Collapse
|
25
|
Mellone M, Gardoni F. Glutamatergic mechanisms in l-DOPA-induced dyskinesia and therapeutic implications. J Neural Transm (Vienna) 2018; 125:1225-1236. [DOI: 10.1007/s00702-018-1846-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/23/2018] [Indexed: 02/01/2023]
|
26
|
Picconi B, Hernández LF, Obeso JA, Calabresi P. Motor complications in Parkinson's disease: Striatal molecular and electrophysiological mechanisms of dyskinesias. Mov Disord 2017; 33:867-876. [PMID: 29219207 DOI: 10.1002/mds.27261] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022] Open
Abstract
Long-term levodopa (l-dopa) treatment in patients with Parkinson´s disease (PD) is associated with the development of motor complications (ie, motor fluctuations and dyskinesias). The principal etiopathogenic factors are the degree of nigro-striatal dopaminergic loss and the duration and dose of l-dopa treatment. In this review article we concentrate on analysis of the mechanisms underlying l-dopa-induced dyskinesias, a phenomenon that causes disability in a proportion of patients and that has not benefited from major therapeutic advances. Thus, we discuss the main neurotransmitters, receptors, and pathways that have been thought to play a role in l-dopa-induced dyskinesias from the perspective of basic neuroscience studies. Some important advances in deciphering the molecular pathways involved in these abnormal movements have occurred in recent years to reveal potential targets that could be used for therapeutic purposes. However, it has not been an easy road because there have been a plethora of components involved in the generation of these undesired movements, even bypassing the traditional and well-accepted dopamine receptor activation, as recently revealed by optogenetics. Here, we attempt to unify the available data with the hope of guiding and fostering future research in the field of striatal activation and abnormal movement generation. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Ledia F Hernández
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Jose A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Paolo Calabresi
- Fondazione Santa Lucia, IRCCS, Rome, Italy.,Clinica Neurologica, Università degli studi di Perugia, Ospedale Santa Maria della Misericordia, Perugia, Italy
| |
Collapse
|
27
|
Deficient striatal adaptation in aminergic and glutamatergic neurotransmission is associated with tardive dyskinesia in non-human primates exposed to antipsychotic drugs. Neuroscience 2017; 361:43-57. [DOI: 10.1016/j.neuroscience.2017.07.068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/10/2017] [Accepted: 07/29/2017] [Indexed: 11/23/2022]
|
28
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|
29
|
Blesa J, Trigo-Damas I, del Rey NLG, Obeso JA. The use of nonhuman primate models to understand processes in Parkinson’s disease. J Neural Transm (Vienna) 2017; 125:325-335. [DOI: 10.1007/s00702-017-1715-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/16/2017] [Indexed: 02/07/2023]
|
30
|
Emmitte KA. mGlu5negative allosteric modulators: a patent review (2013 - 2016). Expert Opin Ther Pat 2017; 27:691-706. [DOI: 10.1080/13543776.2017.1280466] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
31
|
Morin N, Morissette M, Grégoire L, Di Paolo T. mGlu5, Dopamine D2 and Adenosine A2A Receptors in L-DOPA-induced Dyskinesias. Curr Neuropharmacol 2017; 14:481-93. [PMID: 26639458 PMCID: PMC4983750 DOI: 10.2174/1570159x14666151201185652] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 11/04/2015] [Accepted: 11/11/2015] [Indexed: 02/07/2023] Open
Abstract
Patients with Parkinson's disease (PD) receiving L-3,4-dihydroxyphenylalanine (L-DOPA, the gold-standard treatment for this disease) frequently develop abnormal involuntary movements, termed L-DOPA-induced dyskinesias (LID). Glutamate overactivity is well documented in PD and LID. An approach to manage LID is to add to L-DOPA specific agents to reduce dyskinesias such as metabotropic glutamate receptor (mGlu receptor) drugs. This article reviews the contribution of mGlu type 5 (mGlu5) receptors in animal models of PD. Several mGlu5 negative allosteric modulators acutely attenuate LID in 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) monkeys and 6-hydroxydopamine(6-OHDA)-lesioned rats. Chronic administration of mGlu5 negative allosteric modulators to MPTP monkeys and 6-OHDA rats also attenuates LID while maintaining the antiparkinsonian effect of L-DOPA. Radioligand autoradiography shows an elevation of striatal mGlu5 receptors of dyskinetic L-DOPA-treated MPTP monkeys but not in those without LID. The brain molecular correlates of the long-term effect of mGlu5 negative allosteric modulators treatments with L-DOPA attenuating development of LID was shown to extend beyond mGlu5 receptors with normalization of glutamate activity in the basal ganglia of L-DOPA-induced changes of NMDA, AMPA, mGlu2/3 receptors and VGlut2 transporter. In the basal ganglia, mGlu5 receptor negative allosteric modulators also normalize the L-DOPA-induced changes of dopamine D2receptors, their associated signaling proteins (ERK1/2 and Akt/GSK3β) and neuropeptides (preproenkephalin, preprodynorphin) as well as the adenosine A2A receptors expression. These results show in animal models of PD reduction of LID with mGlu5 negative allosteric modulation associated with normalization of glutamate, dopamine and adenosine receptors suggesting a functional link of these receptors in chronic treatment with L-DOPA.
Collapse
Affiliation(s)
| | | | | | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre de recherche du CHU de Québec, 2705 Laurier Boulevard, Quebec, Qc, Canada, G1V 4G2.
| |
Collapse
|
32
|
Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 2017; 115:179-191. [DOI: 10.1016/j.phrs.2016.11.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
33
|
Bruno V, Caraci F, Copani A, Matrisciano F, Nicoletti F, Battaglia G. The impact of metabotropic glutamate receptors into active neurodegenerative processes: A "dark side" in the development of new symptomatic treatments for neurologic and psychiatric disorders. Neuropharmacology 2016; 115:180-192. [PMID: 27140693 DOI: 10.1016/j.neuropharm.2016.04.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/22/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Metabotropic glutamate (mGlu) receptor ligands are under clinical development for the treatment of CNS disorders with high social and economic burden, such as schizophrenia, major depressive disorder (MDD), and Parkinson's disease (PD), and are promising drug candidates for the treatment of Alzheimer's disease (AD). So far, clinical studies have shown symptomatic effects of mGlu receptor ligands, but it is unknown whether these drugs act as disease modifiers or, at the opposite end, they accelerate disease progression by enhancing neurodegeneration. This is a fundamental issue in the treatment of PD and AD, and is also an emerging theme in the treatment of schizophrenia and MDD, in which neurodegeneration is also present and contribute to disease progression. Moving from in vitro data and preclinical studies, we discuss the potential impact of drugs targeting mGlu2, mGlu3, mGlu4 and mGlu5 receptor ligands on active neurodegeneration associated with AD, PD, schizophrenia, and MDD. We wish to highlight that our final comments on the best drug candidates are not influenced by commercial interests or by previous or ongoing collaborations with drug companies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Valeria Bruno
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy.
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; I.R.C.C.S. Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; National Research Council, Institute of Biostructure and Bioimaging (IBB-CNR), 95126 Catania, Italy
| | - Francesco Matrisciano
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy
| | | |
Collapse
|
34
|
Litim N, Morissette M, Di Paolo T. Metabotropic glutamate receptors as therapeutic targets in Parkinson's disease: An update from the last 5 years of research. Neuropharmacology 2016; 115:166-179. [PMID: 27055772 DOI: 10.1016/j.neuropharm.2016.03.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Disturbance of glutamate neurotransmission in Parkinson's disease (PD) and l-DOPA induced dyskinesia (LID) is well documented. This review focuses on advances during the past five years on pharmacological modulation of metabotropic glutamate (mGlu) receptors in relation to anti-parkinsonian activity, LID attenuation, and neuroprotection. Drug design and characterization have led to the development of orthosteric agonists binding the same site as glutamate and Positive and Negative Allosteric modulators (PAMs and NAMs) binding sites different from the orthosteric site and offering subtype selectivity. Inhibition of group I (mGlu1 and mGlu5) receptors with NAMs and activation of group II (mGlu2 and 3 receptors) and group III (mGlu 4, 7 and 8 receptors) with PAMs and orthosteric agonists have shown their potential to inhibit glutamate release and attenuate excitotoxicity. Earlier and recent studies have led to the development of mGlu5 receptors NAMs to reduce LID and for neuroprotection, mGlu3 receptor agonists for neuroprotection while mGlu4 receptor PAMs and agonists for antiparkinsonian effects and neuroprotection. Furthermore, homo- and heterodimers of mGlu receptors are documented and highlight the complexity of the functioning of these receptors. Research on partial allosteric modulators and biased mGlu receptor allosteric modulators offer new glutamatergic drugs with better therapeutic effects and less off target adverse activity. Thus these various mGlu receptor targets will enable the development of novel drugs with improved clinical effects for normalization of glutamate transmission, treat PD and LID relief. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Nadhir Litim
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada; Faculty of Pharmacy, Laval University, Quebec City, Canada
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada; Faculty of Pharmacy, Laval University, Quebec City, Canada.
| |
Collapse
|
35
|
Majláth Z, Török N, Toldi J, Vécsei L. Promising therapeutic agents for the treatment of Parkinson’s disease. Expert Opin Biol Ther 2016; 16:787-99. [DOI: 10.1517/14712598.2016.1164687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
LaCrosse AL, Taylor SB, Nemirovsky NE, Gass JT, Olive MF. mGluR5 Positive and Negative Allosteric Modulators Differentially Affect Dendritic Spine Density and Morphology in the Prefrontal Cortex. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2016; 14:476-85. [PMID: 25921744 DOI: 10.2174/1871527314666150429112849] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 01/17/2015] [Accepted: 01/19/2015] [Indexed: 12/31/2022]
Abstract
Positive and negative allosteric modulators (PAMs and NAMs, respectively) of type 5 metabotropic glutamate receptors (mGluR5) are currently being investigated as novel treatments for neuropsychiatric diseases including drug addiction, schizophrenia, and Fragile X syndrome. However, only a handful of studies have examined the effects of mGluR5 PAMs or NAMs on the structural plasticity of dendritic spines in otherwise naïve animals, particularly in brain regions mediating executive function. In the present study, we assessed dendritic spine density and morphology in pyramidal cells of the medial prefrontal cortex (mPFC) after repeated administration of either the prototypical mGluR5 PAM 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5- yl)benzamide (CDPPB, 20 mg/kg), the clinically utilized mGluR5 NAM 1-(3-chlorophenyl)-3-(3-methyl-5-oxo-4Himidazol- 2-yl)urea (fenobam, 20 mg/kg), or vehicle in male Sprague-Dawley rats. Following once daily treatment for 10 consecutive days, coronal brain sections containing the mPFC underwent diolistic labeling and 3D image analysis of dendritic spines. Compared to vehicle treated animals, rats administered fenobam exhibited significant increases in dendritic spine density and the overall frequency of spines with small (<0.2 μm) head diameters, decreases in frequency of spines with medium (0.2-0.4 μm) head diameters, and had no changes in frequency of spines with large head diameters (>0.4 μm). Administration of CDPPB had no discernable effects on dendritic spine density or morphology, and neither CDPPB nor fenobam had any effect on spine length or volume. We conclude that mGluR5 PAMs and NAMs differentially affect mPFC dendritic spine structural plasticity in otherwise naïve animals, and additional studies assessing their effects in combination with cognitive or behavioral tasks are needed.
Collapse
Affiliation(s)
| | | | | | | | - Michael F Olive
- Department of Psychology, Arizona State University, PO Box 871104, Tempe, AZ 85287, USA.
| |
Collapse
|
37
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 348] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
38
|
Glutamatergic pathways as a target for the treatment of dyskinesias in Parkinson's disease. Biochem Soc Trans 2015; 42:600-4. [PMID: 24646284 DOI: 10.1042/bst20140006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PD (Parkinson's disease) is characterized by some typical motor features that are caused by striatal dopamine depletion and respond well to dopamine-replacement therapy with L-dopa. Unfortunately, the majority of PD patients treated with L-dopa develop abnormal involuntary movements (dyskinesias) within a few years. The mechanisms underlying the development of LIDs (L-dopa-induced dyskinesias) involve, on one hand, a presynaptic dysregulation of dopamine release and clearance and, on the other hand, an abnormal postsynaptic response to dopamine in the brain. There is a large amount of evidence that these dopamine-dependent mechanisms are modulated by glutamatergic pathways and glutamate receptors. The present article summarizes the pathophysiological role of glutamatergic pathways in LID and reviews pre-clinical and clinical results obtained using pharmacological modulators of different classes and subtypes of glutamate receptors to treat parkinsonian dyskinesias.
Collapse
|
39
|
Niccolini F, Rocchi L, Politis M. Molecular imaging of levodopa-induced dyskinesias. Cell Mol Life Sci 2015; 72:2107-17. [PMID: 25681866 PMCID: PMC11113208 DOI: 10.1007/s00018-015-1854-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/06/2015] [Accepted: 02/09/2015] [Indexed: 12/15/2022]
Abstract
Levodopa-induced dyskinesias (LIDs) occur in the majority of patients with Parkinson's disease (PD) following years of levodopa treatment. The pathophysiology underlying LIDs in PD is poorly understood, and current treatments generate only minor benefits for the patients. Studies with positron emission tomography (PET) molecular imaging have demonstrated that in advanced PD patients, levodopa administration induces sharp increases in striatal dopamine levels, which correlate with LIDs severity. Fluctuations in striatal dopamine levels could be the result of the attenuated buffering ability in the dopaminergically denervated striatum. Lines of evidence from PET studies indicate that serotonergic terminals could also be responsible for the development of LIDs in PD by aberrantly processing exogenous levodopa and by releasing dopamine in a dysregulated manner from the serotonergic terminals. Additionally, other downstream mechanisms involving glutamatergic, cannabinoid, opioid, cholinergic, adenosinergic, and noradrenergic systems may contribute in the development of LIDs. In this article, we review the findings from preclinical, clinical, and molecular imaging studies, which have contributed to our understanding the pathophysiology of LIDs in PD.
Collapse
Affiliation(s)
- Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Lorenzo Rocchi
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| |
Collapse
|
40
|
Jourdain VA, Morin N, Grégoire L, Morissette M, Di Paolo T. Changes in glutamate receptors in dyskinetic parkinsonian monkeys after unilateral subthalamotomy. J Neurosurg 2015; 123:1383-93. [PMID: 25932606 DOI: 10.3171/2014.10.jns141570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Unilateral subthalamotomy is a surgical procedure that may be used to alleviate disabling levodopa-induced dyskinesias (LIDs) in patients with Parkinson disease (PD). However, the mechanisms involved in LID remain largely unknown. The subthalamic nucleus (STN) is the sole glutamatergic nucleus within the basal ganglia, and its lesion may produce changes in glutamate receptors in various areas of the basal ganglia. The authors aimed to investigate the biochemical changes in glutamate receptors in striatal and pallidal regions of the basal ganglia after lesion of the STN in parkinsonian macaque monkeys. METHODS The authors treated 12 female ovariectomized monkeys with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce PD-like symptoms, treated 8 of these animals with 3,4-dihydroxy-l-phenylalanine (L-DOPA; levodopa) to induce LID, and performed unilateral subthalamotomy in 4 of these 8 monkeys. Four additional monkeys were treated with saline only and were used as controls. The MPTP monkeys had previously been shown to respond behaviorally to lower doses of levodopa after the STN lesion. Autoradiography of slices from postmortem brain tissues was used to visualize changes in the specific binding of striatal and pallidal ionotropic glutamate receptors (that is, of the α-amino-3-hydroxy 5-methyl-4-isoxazole propionate [AMPA] and N-methyl-d-aspartate [NMDA] NR1/NR2B subunit receptors) and of metabotropic glutamate (mGlu) receptors (that is, mGlu2/3 and mGlu5 receptors). The specific binding and distribution of glutamate receptors in the basal ganglia of the levodopa-treated, STN-lesioned MPTP monkeys were compared with those in the saline-treated control monkeys and in the saline-treated and levodopa-treated MPTP monkeys. RESULTS The autoradiographic results indicated that none of the pharmacological and surgical treatments produced changes in the specific binding of AMPA receptors in the basal ganglia. Levodopa treatment increased the specific binding of NMDA receptors in the basal ganglia. Subthalamotomy reversed these increases in the striatum, but in the globus pallidus (GP), the subthalamotomy reversed these increases only contralaterally. Levodopa treatment reversed MPTP-induced increases in mGlu2/3 receptors only in the GP. mGlu2/3 receptor-specific binding in the striatum and GP decreased bilaterally in the levodopa-treated, STN-lesioned MPTP monkeys compared with the other 3 groups. Compared with mGlu5 receptor-specific binding in the control monkeys, that of the levodopa-treated MPTP monkeys increased in the dorsal putamen and remained unchanged in the caudate nucleus and in the GP. CONCLUSIONS These results implicate glutamate receptors in the previously observed benefits of unilateral subthalamotomy to improve motor control.
Collapse
Affiliation(s)
- Vincent A Jourdain
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Nicolas Morin
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Laurent Grégoire
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| |
Collapse
|
41
|
Plasmalogen precursor analog treatment reduces levodopa-induced dyskinesias in parkinsonian monkeys. Behav Brain Res 2015; 286:328-37. [PMID: 25771209 DOI: 10.1016/j.bbr.2015.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 01/28/2023]
Abstract
L-DOPA-induced dyskinesias (LID) remain a serious obstacle in the treatment of Parkinson's disease (PD). The objective of this study was to test a new target for treatment of dyskinesias, ethanolamine plasmalogens (PlsEtn). PlsEtn play critical roles in membrane structure mediated functions and as a storage depot of polyunsaturated fatty acids such as docosahexaenoic acid (DHA, omega-3) known to reduce dyskinesias. The motor effect of a daily treatment for 12 days of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Macaca fascicularis monkeys with DHA (100mg/kg) was compared to the DHA-PlsEtn precursor PPI-1011 (50mg/kg). PPI-1011 and DHA reduced LID while maintaining the antiparkinsonian activity of l-DOPA, however the PPI-1011 effect was observed at the first behavioral time point analyzed following drug administration (day 2) whereas the effect of DHA was not observed until after 10 days of administration. DHA treatment increased plasma DHA levels 2-3× whereas PPI-1011 had no effect. DHA and PPI-1011 increased DHA-PlsEtn levels by 1.5-2× while DHA-phosphatidylethanolamine (PtdEtn) levels remained unaffected. DHA treatment also elevated very long chain fatty acid containing PtdEtn and reduced non-DHA containing PtdEtn and PlsEtn levels. PPI-1011 had no effect on these systems. LID scores were inversely correlated with serum DHA-PlsEtn/total PlsEtn ratios levels in DHA and PPI-1011 treated monkeys. Hence, the antidyskinetic activity of DHA and PPI-1011 in MPTP monkeys appears to be associated with the increase of serum DHA-PlsEtn concentrations. This is the first study reporting an antidyskinetic response to augmentation of DHA-PlsEtn using a plasmalogen precursor thus providing a novel drug target for dyskinesias.
Collapse
|
42
|
Neurobiology of l-DOPA induced dyskinesia and the novel therapeutic strategies. Biomed Pharmacother 2015; 70:283-93. [DOI: 10.1016/j.biopha.2015.01.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 01/23/2015] [Indexed: 12/27/2022] Open
|
43
|
Fox SH, Brotchie JM, Johnston TM. Primate Models of Complications Related to Parkinson Disease Treatment. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
44
|
Schaeffer E, Pilotto A, Berg D. Pharmacological strategies for the management of levodopa-induced dyskinesia in patients with Parkinson's disease. CNS Drugs 2014; 28:1155-84. [PMID: 25342080 DOI: 10.1007/s40263-014-0205-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
L-Dopa-induced dyskinesias (LID) are the most common adverse effects of long-term dopaminergic therapy in Parkinson's disease (PD). However, the exact mechanisms underlying dyskinesia are still unclear. For a long time, nigrostriatal degeneration and pulsatile stimulation of striatal postsynaptic receptors have been highlighted as the key factors for the development of LID. In recent years, PD models have revealed a wide range of non-dopaminergic neurotransmitter systems involved in pre- and postsynaptic changes and thereby contributing to the pathophysiology of LID. In the current review, we focus on therapeutic LID targets, mainly based on agents acting on dopaminergic, glutamatergic, serotoninergic, adrenergic, and cholinergic systems. Despite a large number of clinical trials, currently only amantadine and, to a lesser extent, clozapine are being used as effective strategies in the treatment of LID in clinical settings. Thus, in the second part of the article, we review the placebo-controlled trials on LID treatment in order to disentangle the changing scenario of drug development. Promising results include the extension of L-dopa action without inducing LID of the novel monoamine oxidase B- and glutamate-release inhibitor safinamide; however, this had no obvious effect on existing LID. Others, like the metabotropic glutamate-receptor antagonist AFQ056, showed promising results in some of the studies; however, confirmation is still lacking. Thus, to date, strategies of continuous dopaminergic stimulation seem the most promising to prevent or ameliorate LID. The success of future therapeutic strategies once moderate to severe LID occur will depend on the translation from preclinical experimental models into clinical practice in a bidirectional process.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tuebingen, Hoppe Seyler-Strasse 3, 72076, Tübingen, Germany
| | | | | |
Collapse
|
45
|
Rascol O, Fox S, Gasparini F, Kenney C, Di Paolo T, Gomez-Mancilla B. Use of metabotropic glutamate 5-receptor antagonists for treatment of levodopa-induced dyskinesias. Parkinsonism Relat Disord 2014; 20:947-56. [DOI: 10.1016/j.parkreldis.2014.05.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/02/2014] [Accepted: 05/02/2014] [Indexed: 10/25/2022]
|
46
|
Morin N, Di Paolo T. Pharmacological Treatments Inhibiting Levodopa-Induced Dyskinesias in MPTP-Lesioned Monkeys: Brain Glutamate Biochemical Correlates. Front Neurol 2014; 5:144. [PMID: 25140165 PMCID: PMC4122180 DOI: 10.3389/fneur.2014.00144] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/18/2014] [Indexed: 12/21/2022] Open
Abstract
Anti-glutamatergic drugs can relieve Parkinson’s disease (PD) symptoms and decrease l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesias (LID). This review reports relevant studies investigating glutamate receptor subtypes in relation to motor complications in PD patients and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys. Antagonists of the ionotropic glutamate receptors, such as N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, display antidyskinetic activity in PD patients and animal models such as the MPTP monkey. Metabotropic glutamate 5 (mGlu5) receptor antagonists were shown to reduce the severity of LID in PD patients as well as in already dyskinetic non-human primates and to prevent the development of LID in de novo treatments in non-human primates. An increase in striatal post-synaptic NMDA, AMPA, and mGlu5 receptors is documented in PD patients and MPTP monkeys with LID. This increase can be prevented in MPTP monkeys with the addition of a specific glutamate receptor antagonist to the l-DOPA treatment and also with drugs of various pharmacological specificities suggesting multiple receptor interactions. This is yet to be well documented for presynaptic mGlu4 and mGlu2/3 and offers additional new promising avenues.
Collapse
Affiliation(s)
- Nicolas Morin
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec , Quebec City, QC , Canada ; Faculty of Pharmacy, Laval University , Quebec City, QC , Canada
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec , Quebec City, QC , Canada ; Faculty of Pharmacy, Laval University , Quebec City, QC , Canada
| |
Collapse
|
47
|
Harikrishna Reddy D, Misra S, Medhi B. Advances in Drug Development for Parkinson's Disease: Present Status. Pharmacology 2014; 93:260-71. [DOI: 10.1159/000362419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/24/2014] [Indexed: 11/19/2022]
|
48
|
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: 2.9] [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
|
49
|
Modeling dyskinesia in animal models of Parkinson disease. Exp Neurol 2014; 256:105-16. [DOI: 10.1016/j.expneurol.2013.01.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/12/2013] [Accepted: 01/21/2013] [Indexed: 01/23/2023]
|
50
|
Bezard E, Pioli EY, Li Q, Girard F, Mutel V, Keywood C, Tison F, Rascol O, Poli SM. The mGluR5 negative allosteric modulator dipraglurant reduces dyskinesia in the MPTP macaque model. Mov Disord 2014; 29:1074-9. [DOI: 10.1002/mds.25920] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/08/2014] [Accepted: 04/21/2014] [Indexed: 11/08/2022] Open
Affiliation(s)
- Erwan Bezard
- Motac neuroscience; Manchester UK
- Université de Bordeaux; Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux France
- CNRS; Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux France
- Service de Neurologie; CHU de Bordeaux; Pessac France
- Institute of Laboratory Animal Sciences; China Academy of Medical Sciences; Beijing China
| | | | - Qin Li
- Motac neuroscience; Manchester UK
- Institute of Laboratory Animal Sciences; China Academy of Medical Sciences; Beijing China
| | | | | | | | - Francois Tison
- Université de Bordeaux; Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux France
- CNRS; Institut des Maladies Neurodégénératives, UMR 5293; Bordeaux France
- Service de Neurologie; CHU de Bordeaux; Pessac France
| | - Olivier Rascol
- CIC Toulouse; France
- Départements de Pharmacologie Clinique et Neurosciences; INSERM CIC9302; CHU de Toulouse France
- Service de Pharmacologie; Faculté de Médecine, CHU de Toulouse, Université de Toulouse; France
| | | |
Collapse
|