1
|
Delmas CVL, Munro J, Bérard M, Di Paolo T, Morissette M, Tremblay ME, Parent A, Parent M. Serotonin innervation of the subthalamic nucleus in parkinsonian monkeys. Neurobiol Dis 2025; 211:106938. [PMID: 40320179 DOI: 10.1016/j.nbd.2025.106938] [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: 02/23/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
The subthalamic nucleus (STN), the main driving force of the basal ganglia, is innervated by brainstem serotonin (5-HT) neurons with highly plastic axonal arborization. A pathologically-induced rearrangement of the ascending 5-HT projections could contribute to the disrupted firing pattern of STN neurons observed in Parkinson's disease (PD). This light and electron microscope study was designed to characterize the neuroadaptive changes of 5-HT inputs to the different functional territories of the STN in four cynomolgus monkeys (Macaca fascicularis) rendered parkinsonian by systemic injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and four control animals. Using an unbiased stereological approach, we report a significant decrease of the density of 5-HT axon varicosities immunolabeled for the 5-HT membrane transporter (SERT), across all STN functional territories of MPTP-treated monkeys. In MPTP-treated animals, the SERT+ axon varicosities are larger than in control monkeys. In both experimental conditions they are only partially synaptic. A preserved length of 5-HT axons in the STN along with a conserved number of 5-HT neurons in the dorsal raphe nucleus is observed. Overall, our results indicate that, in parkinsonian monkeys, the 5-HT axons projecting to the STN are preserved but endowed with significantly less axon varicosities. Such neuroadaptive change could lead to a lower ambient level of 5-HT in this basal ganglia component, representing a compensatory mechanism designed to cope with the hyperexcitability of STN neurons that is known to occur in PD.
Collapse
Affiliation(s)
- C V L Delmas
- CERVO Brain Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - J Munro
- CERVO Brain Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - M Bérard
- CERVO Brain Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - T Di Paolo
- Centre de recherche du CHU de Québec and Faculty of Pharmacy, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - M Morissette
- Centre de recherche du CHU de Québec and Faculty of Pharmacy, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - M E Tremblay
- University of Victoria, Division of Medical Sciences, Victoria, BC V8P 5C2, Canada
| | - A Parent
- CERVO Brain Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - M Parent
- CERVO Brain Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada.
| |
Collapse
|
2
|
Dunker C, Schlegel K, Junker A. Phenol (bio)isosteres in drug design and development. Arch Pharm (Weinheim) 2025; 358:e2400700. [PMID: 39580699 PMCID: PMC11726161 DOI: 10.1002/ardp.202400700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
Due to their versatile properties, phenolic compounds are integral to various biologically active molecules, including many pharmaceuticals. However, their application in drug design is often hindered by issues such as poor oral bioavailability, rapid metabolism, and potential toxicity. This review explores the use of phenol bioisosteres-structurally similar compounds that can mimic the biological activity of phenols while potentially offering improved drug-like properties. We provide an extensive analysis of various phenol bioisosteres, including benzimidazolones, benzoxazolones, indoles, quinolinones, and pyridones, highlighting their impact on the pharmacokinetic and pharmacodynamic profiles of drugs. Case studies illustrate the successful application of these bioisosteres in enhancing metabolic stability, receptor selectivity, and overall therapeutic efficacy. Additionally, the review addresses the challenges associated with phenol bioisosterism, such as maintaining potency and avoiding undesirable side effects. By offering a detailed examination of current strategies and potential future directions, this review serves as a valuable resource for medicinal chemists seeking to optimize phenolic scaffolds in drug development. The insights provided herein aim to facilitate the design of more effective and safer therapeutic agents through strategic bioisosteric modifications.
Collapse
Affiliation(s)
- Calvin Dunker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| | - Katja Schlegel
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| |
Collapse
|
3
|
Gouriou E, Bourque M, Schneider C, Di Paolo T. Exploring Magnetic and Electrical Brain Stimulation in Parkinsonian Dyskinetic Monkeys. Can J Neurol Sci 2024:1-12. [PMID: 39530289 DOI: 10.1017/cjn.2024.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Parkinson's disease (PD) chronic L-Dopa treatment often triggers motor complications, such as L-Dopa-induced dyskinesias (LID). LID are reported to be associated with abnormal glutamatergic activity between the striatum and primary motor cortex (M1), resulting in M1 hyperactivation. Beneficial noninvasive brain stimulation (NIBS) paradigms were reported to normalize glutamatergic activity. The objective of the present study was thus to set up a NIBS paradigm in parkinsonian monkeys to investigate motor behavior under basal conditions and with L-Dopa treatment-inducing dyskinesias. METHODS Motor behavior was investigated in five 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) dyskinetic female Macaca fascicularis monkey models of PD, allowing us to monitor the administration of NIBS and drugs. NIBS used were inhibitory protocols, that is, cathodal transcranial direct current stimulation (c-tDCS) and continuous theta-burst stimulation (cTBS). A procedure of three weeks was developed to progressively acclimate animals to the experimental conditions, equipment and noise of c-tDCS and cTBS before stimulating them with either vehicle or L-Dopa. RESULTS One session of c-tDCS with L-Dopa yielded no effect, whereas five sessions briefly reduced LID but decreased the duration of L-Dopa anti-PD effects. cTBS alone improved (decreased) parkinsonian scores as compared to sham stimulation or vehicle alone. Two sessions of cTBS with L-Dopa decreased LID without affecting L-Dopa anti-PD effects. CONCLUSION This is the first study testing c-tDCS and cTBS on the motor behavior of MPTP dyskinetic monkeys. As compared to medicated patients, MPTP monkeys offer the opportunity to evaluate NIBS after-effects in drug-free and LID conditions, which are critical in the search for new PD treatment.
Collapse
Affiliation(s)
- Estelle Gouriou
- Noninvasive Neurostimulation Laboratory, Neuroscience Unit, Research Center of CHU de Québec - Université Laval, Quebec, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculty of Medicine, Université Laval, Quebec, Canada
| | - Mélanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
| | - Cyril Schneider
- Noninvasive Neurostimulation Laboratory, Neuroscience Unit, Research Center of CHU de Québec - Université Laval, Quebec, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculty of Medicine, Université Laval, Quebec, Canada
- Faculty of Medicine, School of Rehabilitation Science, Université Laval, Quebec, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculty of Pharmacy, Université Laval, Quebec, Canada
| |
Collapse
|
4
|
Dos Santos Pereira M, Dias de Abreu GH, Vanderlei LCA, Raisman-Vozari R, Guimarães FS, Lu HC, Michel PP, Del Bel E. 4'-fluorocannabidiol associated with capsazepine restrains L-DOPA-induced dyskinesia in hemiparkinsonian mice: Contribution of anti-inflammatory and anti-glutamatergic mechanisms. Neuropharmacology 2024; 251:109926. [PMID: 38554815 PMCID: PMC11988267 DOI: 10.1016/j.neuropharm.2024.109926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024]
Abstract
We tested the efficacy of 4'-fluorocannabidiol (4'-F-CBD), a semisynthetic cannabidiol derivative, and HU-910, a cannabinoid receptor 2 (CB2) agonist in resolving l-DOPA-induced dyskinesia (LID). Specifically, we were interested in studying whether these compounds could restrain striatal inflammatory responses and rescue glutamatergic disturbances characteristic of the dyskinetic state. C57BL/6 mice were rendered hemiparkinsonian by unilateral striatal lesioning with 6-OHDA. Abnormal involuntary movements were then induced by repeated i.p. injections of l-DOPA + benserazide. After LID was installed, the effects of a 3-day treatment with 4'-F-CBD or HU-910 in combination or not with the TRPV1 antagonist capsazepine (CPZ) or CB2 agonists HU-308 and JWH015 were assessed. Immunostaining was conducted to investigate the impacts of 4'-F-CBD and HU-910 (with CPZ) on inflammation and glutamatergic synapses. Our results showed that the combination of 4'-F-CBD + CPZ, but not when administered alone, decreased LID. Neither HU-910 alone nor HU-910+CPZ were effective. The CB2 agonists HU-308 and JWH015 were also ineffective in decreasing LID. Both combination treatments efficiently reduced microglial and astrocyte activation in the dorsal striatum of dyskinetic mice. However, only 4'-F-CBD + CPZ normalized the density of glutamate vesicular transporter-1 (vGluT1) puncta colocalized with the postsynaptic density marker PSD95. These findings suggest that 4'-F-CBD + CPZ normalizes dysregulated cortico-striatal glutamatergic inputs, which could be involved in their anti-dyskinetic effects. Although it is not possible to rule out the involvement of anti-inflammatory mechanisms, the decrease in striatal neuroinflammation markers by 4'-F-CBD and HU-910 without an associated reduction in LID indicates that they are insufficient per se to prevent LID manifestations.
Collapse
Affiliation(s)
- Maurício Dos Santos Pereira
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil; Paris Brain Institute, Inserm, CNRS, Sorbonne Université, Paris, France.
| | - Gabriel Henrique Dias de Abreu
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Bimolecular Sciences, Indiana University, Bloomington, United States.
| | | | | | | | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Bimolecular Sciences, Indiana University, Bloomington, United States.
| | | | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
5
|
Cervetto C, Maura G, Guidolin D, Amato S, Ceccoli C, Agnati LF, Marcoli M. Striatal astrocytic A2A-D2 receptor-receptor interactions and their role in neuropsychiatric disorders. Neuropharmacology 2023:109636. [PMID: 37321323 DOI: 10.1016/j.neuropharm.2023.109636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Italy.
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Cristina Ceccoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Luigi F Agnati
- Department of Biochemical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy; Center of Excellence for Biomedical Research, University of Genova, Italy.
| |
Collapse
|
6
|
Herrero MT, Yuste JE, Cuenca-Bermejo L, Almela P, Arenas-Betancur L, De Pablos V, Gonzalez-Cuello A, Del Bel E, Navarro-Zaragoza J, Fernández-Villalba E. 7-Nitroindazole reduces L-DOPA-induced dyskinesias in non-human Parkinsonian primate. Open Biol 2023; 13:220370. [PMID: 37192671 DOI: 10.1098/rsob.220370] [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: 12/19/2022] [Accepted: 03/31/2023] [Indexed: 05/18/2023] Open
Abstract
Nitric oxide (NO) plays a pivotal role in integrating dopamine transmission in the basal ganglia and has been implicated in the pathogenesis of Parkinson disease (PD). The objective of this study was to ascertain whether the NO synthase inhibitor, 7-nitroindazole (7-NI), is able to reduce L-DOPA-induced dyskinesias (LIDs) in a non-human primate model of PD chronically intoxicated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Six Parkinsonian macaques were treated daily with L-DOPA for 3-4 months until they developed LIDs. Three animals were then co-treated with a single dose of 7-NI administered 45 min before each L-DOPA treatment. Dyskinetic MPTP-treated monkeys showed a significant decrease in LIDs compared with their scores without 7-NI treatment (p < 0.05). The anti-Parkinsonian effect of L-DOPA was similar in all three monkeys with and without 7-NI co-treatment. This improvement was significant with respect to the intensity and duration of LIDs while the beneficial effect of L-DOPA treatment was maintained and could represent a promising therapy to improve the quality of life of PD patients.
Collapse
Affiliation(s)
- M T Herrero
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - J E Yuste
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
| | - L Cuenca-Bermejo
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - P Almela
- Department of Pharmacology, School of Medicine, University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - L Arenas-Betancur
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
| | - V De Pablos
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - A Gonzalez-Cuello
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - E Del Bel
- Department of Basic and Oral Biology, Faculty of Odontology of Ribeirão Preto (FORP-USP) and Center for Research Support on Applied Neuroscience (NAPNA-USP), University of São Paulo, Ribeirão Preto, SP 14040-904, Brazil
| | - J Navarro-Zaragoza
- Department of Pharmacology, School of Medicine, University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - E Fernández-Villalba
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
7
|
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
|
8
|
Prasher P, Mall T, Sharma M. Cyclic carbamates in medicine: A clinical perspective. Drug Dev Res 2023; 84:397-405. [PMID: 36651662 DOI: 10.1002/ddr.22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023]
Abstract
Carbamate group is mainly used for designing prodrugs to achieve first-pass and systemic stability against enzyme hydrolysis as the carbamate functionality is recognized by esterase enzymes. As compared to the ester functionality, the carbamate group shows a lesser lability towards enzyme hydrolysis, but a higher susceptibility than amides. Cyclic carbamates present a unique motif in the contemporary drug discovery and development owing to the presence of a polar, and sterically small, constrained Hydrogen-bonding acceptor atom. The metabolic stability of 5/6-membered cyclic carbamates are higher as compared to their acyclic counterparts as the former do not undergo metabolic ring opening under physiological conditions. Besides, the metabolic lability of acyclic carbamates is determined by the degree of substitution at the endocyclic/exocyclic "N" atom, which further enables the design and development of various carbamate drugs or prodrugs. As such, the metabolic stability of carbamates follows the order: Cyclic carbamates > Alkyl-OCO-NH2 » Alkyl-OCO-NHAcyl ∼ Alkyl-OCO-NHAryl ≥ Aryl-OCO-N(endocyclic) ∼ Aryl-OCO-N(Alkyl)2 ≥ Alkyl-OCO-N(endocyclic) ≥ Alkyl-OCO-N(Alkyl)2 ∼ Alkyl-OCO-NHAlkyl » Aryl-OCO-NHAlkyl.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Tanisqa Mall
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
9
|
Bourque M, Grégoire L, Patel W, Dickens D, Snodgrass R, Di Paolo T. AV-101, a Pro-Drug Antagonist at the NMDA Receptor Glycine Site, Reduces L-Dopa Induced Dyskinesias in MPTP Monkeys. Cells 2022; 11:cells11223530. [PMID: 36428960 PMCID: PMC9688762 DOI: 10.3390/cells11223530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors have been implicated in L-Dopa-induced dyskinesias (LID) in Parkinson's disease patients, but the use of antagonists that directly inhibit this receptor is associated with severe side effects. L-4-chlorokynurenine (4-Cl-KYN or AV-101) is a pro-drug of 7-chlorokynurenic acid (7-Cl-KYNA), a potent and specific antagonist of the glycine (GlyB) co-agonist site of NMDA receptors. The 7-Cl-KYNA has limited ability to cross the blood-brain barrier, whereas AV-101 readily accesses the brain. We investigated if AV-101 reduces LID in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys while maintaining the antiparkinsonian activity of L-Dopa. A first pilot study using three dyskinetic MPTP monkeys showed that acute AV-101 treatment (250 and 450 mg/kg) reduced LID and maintained the antiparkinsonian activity of L-Dopa. The main study using six additional dyskinetic MPTP monkeys showed that repeated AV-101 treatment (250 mg/kg, b.i.d. for 4 consecutive days) maintained their L-Dopa antiparkinsonian response. We measured significantly less LID when AV-101 was combined with L-Dopa treatment. AV-101 alone or with L-Dopa had no non-motor adverse effects in MPTP monkeys. Our study showed antidyskinetic activity of AV-101 in MPTP monkeys was comparable to amantadine tested previously in our laboratory in this model. We observed no adverse effects with AV-101, which is an improvement over amantadine, with its known side effects.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | - Laurent Grégoire
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | - Waseema Patel
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - David Dickens
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - Ralph Snodgrass
- Vistagen Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC G1V0A6, Canada
- Correspondence:
| |
Collapse
|
10
|
Bandopadhyay R, Mishra N, Rana R, Kaur G, Ghoneim MM, Alshehri S, Mustafa G, Ahmad J, Alhakamy NA, Mishra A. Molecular Mechanisms and Therapeutic Strategies for Levodopa-Induced Dyskinesia in Parkinson's Disease: A Perspective Through Preclinical and Clinical Evidence. Front Pharmacol 2022; 13:805388. [PMID: 35462934 PMCID: PMC9021725 DOI: 10.3389/fphar.2022.805388] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the second leading neurodegenerative disease that is characterized by severe locomotor abnormalities. Levodopa (L-DOPA) treatment has been considered a mainstay for the management of PD; however, its prolonged treatment is often associated with abnormal involuntary movements and results in L-DOPA-induced dyskinesia (LID). Although LID is encountered after chronic administration of L-DOPA, the appearance of dyskinesia after weeks or months of the L-DOPA treatment has complicated our understanding of its pathogenesis. Pathophysiology of LID is mainly associated with alteration of direct and indirect pathways of the cortico-basal ganglia-thalamic loop, which regulates normal fine motor movements. Hypersensitivity of dopamine receptors has been involved in the development of LID; moreover, these symptoms are worsened by concurrent non-dopaminergic innervations including glutamatergic, serotonergic, and peptidergic neurotransmission. The present study is focused on discussing the recent updates in molecular mechanisms and therapeutic approaches for the effective management of LID in PD patients.
Collapse
Affiliation(s)
- Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Nainshi Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ruhi Rana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Gagandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gulam Mustafa
- College of Pharmacy (Boys), Al-Dawadmi Campus, Shaqra University, Riyadh, Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Nabil. A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)—Guwahati, Guwahati, India
| |
Collapse
|
11
|
Dos Santos Pereira M, Abreu GHD, Rocca J, Hamadat S, Raisman-Vozari R, Michel PP, Del Bel E. Contributive Role of TNF-α to L-DOPA-Induced Dyskinesia in a Unilateral 6-OHDA Lesion Model of Parkinson's Disease. Front Pharmacol 2021; 11:617085. [PMID: 33510643 PMCID: PMC7836015 DOI: 10.3389/fphar.2020.617085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Our present objective was to better characterize the mechanisms that regulate striatal neuroinflammation in mice developing L-DOPA-induced dyskinesia (LID). For that, we used 6-hydroxydopamine (6-OHDA)-lesioned mice rendered dyskinetic by repeated intraperitoneal injections of 3,4-dihydroxyphenyl-L-alanine (L-DOPA) and quantified ensuing neuroinflammatory changes in the dopamine-denervated dorsal striatum. LID development was associated with a prominent astrocytic response, and a more moderate microglial cell reaction restricted to this striatal area. The glial response was associated with elevations in two pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β. Treatment with the phytocannabinoid cannabidiol and the transient receptor potential vanilloid-1 (TRPV-1) channel antagonist capsazepine diminished LID intensity and decreased TNF-α levels without impacting other inflammation markers. To possibly reproduce the neuroinflammatory component of LID, we exposed astrocyte and microglial cells in culture to candidate molecules that might operate as inflammatory cues during LID development, i.e., L-DOPA, dopamine, or glutamate. Neither L-DOPA nor dopamine produced an inflammatory response in glial cell cultures. However, glutamate enhanced TNF-α secretion and GFAP expression in astrocyte cultures and promoted Iba-1 expression in microglial cultures. Of interest, the antidyskinetic treatment with cannabidiol + capsazepine reduced TNF-α release in glutamate-activated astrocytes. TNF-α, on its own, promoted the synaptic release of glutamate in cortical neuronal cultures, whereas cannabidiol + capsazepine prevented this effect. Therefore, we may assume that the release of TNF-α by glutamate-activated astrocytes may contribute to LID by exacerbating corticostriatal glutamatergic inputs excitability and maintaining astrocytes in an activated state through a self-reinforcing mechanism.
Collapse
Affiliation(s)
- Maurício Dos Santos Pereira
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,Department of Physiology, FMRP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil.,Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Gabriel Henrique Dias Abreu
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil
| | - Jeremy Rocca
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Sabah Hamadat
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Patrick Pierre Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,Department of Physiology, FMRP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil
| |
Collapse
|
12
|
Pinna A, Serra M, Marongiu J, Morelli M. Pharmacological interactions between adenosine A 2A receptor antagonists and different neurotransmitter systems. Parkinsonism Relat Disord 2020; 80 Suppl 1:S37-S44. [PMID: 33349579 DOI: 10.1016/j.parkreldis.2020.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/23/2020] [Accepted: 10/11/2020] [Indexed: 12/21/2022]
Abstract
While Parkinson's disease (PD) is traditionally characterized by dopaminergic neuron degeneration, several neurotransmitters and neuromodulators besides dopamine are also involved in the onset and progression of the disease and its symptoms. The other principal neurotransmitters/neuromodulators known to control basal ganglia functions and, in particular, motor functions, are GABA, glutamate, serotonin (5-HT), noradrenaline, acetylcholine, adenosine and endocannabinoids. Among these, adenosine is the most relevant, acting through its adenosine A2A receptor. Work in experimental models of PD has established the effects of A2A receptor antagonists, including the alleviation of disrupted dopamine functions and improved efficacy of dopamine replacement therapy. Moreover, positive interactions between A2A receptor antagonists and both D2 and D1 receptor agonists have been described in vitro at the receptor-receptor level or in more complex in vivo models of PD, respectively. In addition, the interactions between A2A receptor antagonists and glutamate ionotropic GluN2B-containing N-Methyl-d-aspartic acid receptors, or metabotropic glutamate (mGlu) receptors, including both mGlu5 receptor inhibitors and mGlu4 receptor activators, have been reported in both in vitro and in vivo animal models of PD, as have positive interactions between A2A and endocannabinoid CB1 receptor antagonists. At the same time, a combination of A2A receptor antagonists and 5-HT1A-5-HT1B receptor agonists have been described to modulate the expression of dyskinesia induced by chronic dopamine replacement therapy.
Collapse
Affiliation(s)
- Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute - Cagliari, Cagliari, Italy.
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- National Research Council of Italy, Neuroscience Institute - Cagliari, Cagliari, Italy; Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
13
|
Lecours C, St-Pierre MK, Picard K, Bordeleau M, Bourque M, Awogbindin IO, Benadjal A, Ibanez FG, Gagnon D, Cantin L, Parent M, Di Paolo T, Tremblay ME. Levodopa partially rescues microglial numerical, morphological, and phagolysosomal alterations in a monkey model of Parkinson's disease. Brain Behav Immun 2020; 90:81-96. [PMID: 32755645 DOI: 10.1016/j.bbi.2020.07.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder. The mechanisms underlying the onset and progression of Levodopa (L-Dopa)-induced dyskinesia (LID) during PD treatment remain elusive. Emerging evidence implicates functional modification of microglia in the development of LID. Thus, understanding the link between microglia and the development of LID may provide the knowledge required to preserve or promote beneficial microglial functions, even during a prolonged L-Dopa treatment. To provide novel insights into microglial functional alterations in PD pathophysiology, we characterized their density, morphology, ultrastructure, and degradation activity in the sensorimotor functional territory of the putamen, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) cynomolgus monkeys. A subset of MPTP monkeys was treated orally with L-Dopa and developed LID similar to PD patients. Using a combination of light, confocal and transmission electron microscopy, our quantitative analyses revealed alterations of microglial density, morphology and phagolysosomal activity following MPTP intoxication that were partially normalized with L-Dopa treatment. In particular, microglial density, cell body and arborization areas were increased in the MPTP monkeys, whereas L-Dopa-treated MPTP animals presented a microglial phenotype similar to the control animals. At the ultrastructural level, microglia did not differ between groups in their markers of cellular stress or aging. Nevertheless, microglia from the MPTP monkeys displayed reduced numbers of endosomes, compared with control animals, that remained lower after L-Dopa treatment. Microglia from MPTP monkeys treated with L-Dopa also had increased numbers of primary lysosomes compared with non-treated MPTP animals, while secondary and tertiary lysosomes remained unchanged. Moreover, a decrease microglial immunoreactivity for CD68, considered a marker of phagocytosis and lysosomal activity, was measured in the MPTP monkeys treated with L-Dopa, compared with non-treated MPTP animals. Taken together, these findings revealed significant changes in microglia during PD pathophysiology that were partially rescued by L-Dopa treatment. Albeit, this L-Dopa treatment conferred phagolysosomal insufficiency on microglia in the dyskinetic Parkinsonian monkeys.
Collapse
Affiliation(s)
- Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Maude Bordeleau
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Integrated Program of Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Melanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Ifeoluwa Oluleke Awogbindin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Amin Benadjal
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Biologie Intégrative et Physiologie, Sorbonne Université, Paris VI, France
| | | | - Dave Gagnon
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada; CERVO Brain Research Center, Québec, QC, Canada
| | - Leo Cantin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada; CERVO Brain Research Center, Québec, QC, Canada
| | - Therese Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Boros F, Vécsei L. Progress in the development of kynurenine and quinoline-3-carboxamide-derived drugs. Expert Opin Investig Drugs 2020; 29:1223-1247. [DOI: 10.1080/13543784.2020.1813716] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Fanni Boros
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences and the University of Szeged, Szeged, Hungary
- Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
15
|
AlShimemeri S, Fox SH, Visanji NP. Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update. Expert Opin Emerg Drugs 2020; 25:131-144. [DOI: 10.1080/14728214.2020.1763954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sohaila AlShimemeri
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Susan H Fox
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
16
|
Receptor Ligands as Helping Hands to L-DOPA in the Treatment of Parkinson's Disease. Biomolecules 2019; 9:biom9040142. [PMID: 30970612 PMCID: PMC6523988 DOI: 10.3390/biom9040142] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/12/2022] Open
Abstract
Levodopa (LD) is the most effective drug in the treatment of Parkinson’s disease (PD). However, although it represents the “gold standard” of PD therapy, LD can cause side effects, including gastrointestinal and cardiovascular symptoms as well as transient elevated liver enzyme levels. Moreover, LD therapy leads to LD-induced dyskinesia (LID), a disabling motor complication that represents a major challenge for the clinical neurologist. Due to the many limitations associated with LD therapeutic use, other dopaminergic and non-dopaminergic drugs are being developed to optimize the treatment response. This review focuses on recent investigations about non-dopaminergic central nervous system (CNS) receptor ligands that have been identified to have therapeutic potential for the treatment of motor and non-motor symptoms of PD. In a different way, such agents may contribute to extending LD response and/or ameliorate LD-induced side effects.
Collapse
|
17
|
Fox SH, Brotchie JM. Viewpoint: Developing drugs for levodopa-induced dyskinesia in PD: Lessons learnt, what does the future hold? Eur J Neurosci 2018; 49:399-409. [PMID: 30269407 DOI: 10.1111/ejn.14173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 12/21/2022]
Abstract
The drive to develop drugs to treat PD starts and ends with the patient. Herein, we discuss how the experience with drug development for LID has led the field in translational studies in PD with advancing ground-breaking science via rigorous clinical trial design, to deliver clinical proof-of-concepts across multiple therapeutic targets. However, issues remain in advancing drugs efficacious preclinically to the clinic, and future studies need to learn from past successes and failures. Such lessons include implementing better early indicators of tolerability, for instance evaluating non-motor symptoms in preclinical models; improving patient-related outcome measures in clinical trials, as well as considering the unique nature of dyskinesia in an individual patient. The field of translational studies needs to become more patient focused to improve successful outcomes.
Collapse
Affiliation(s)
- Susan H Fox
- The Edmond J Safra Program in Parkinson Disease and Movement Disorder Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jonathan M Brotchie
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada.,Atuka Inc, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Modulation of CaMKIIa-GluN2B interaction in levodopa-induced dyskinesia in 6-OHDA-lesioned Parkinson's rats. Biomed Pharmacother 2018; 107:769-776. [PMID: 30142538 DOI: 10.1016/j.biopha.2018.08.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/11/2018] [Accepted: 08/15/2018] [Indexed: 11/23/2022] Open
Abstract
Long-term treatment with L-dopa leads to involuntary aimless movements called L-dopa-induced dyskinesia (LID) has hindered its use in Parkinson's disease (PD) patients. Emerging evidence suggests a possible role of CaMKIIa and its interacting partners in the development of LID. In this study, we found that CaMKIIa was found to form complexes with GluN2B after chronic administration of L-dopa in adult rat striatal neurons. Intrastriatal injection of KN-93 significantly reduced the level of GluN2B in CaMKIIa precipitates with a dose dependent response, as well as reduced the Global ALO AIM score without ablation of the therapeutic response to L-dopa. In parallel, intrastriatal injection of MK-801 significantly alleviated the level of CaMKIIa in GluN2B precipitates compared to LID group (p < 0.01), and this is accompanied by realizing improvement of the Global ALO AIM score also without affect the efficacy of L-dopa. In summary, the present study indicated that CaMKIIa-GluN2B interaction had an important role in the development of LID. Disrupt of this link by intrastriatal infusion of KN-93 or MK-801 ameliorated dyskinesia in 6-OHDA-lesioned PD rats.
Collapse
|
19
|
Cervetto C, Venturini A, Guidolin D, Maura G, Passalacqua M, Tacchetti C, Cortelli P, Genedani S, Candiani S, Ramoino P, Pelassa S, Marcoli M, Agnati LF. Homocysteine and A2A-D2 Receptor-Receptor Interaction at Striatal Astrocyte Processes. J Mol Neurosci 2018; 65:456-466. [PMID: 30030763 DOI: 10.1007/s12031-018-1120-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/11/2018] [Indexed: 01/03/2023]
Abstract
The interaction between adenosine A2A and dopamine D2 receptors in striatal neurons is a well-established phenomenon and has opened up new perspectives on the molecular mechanisms involved in Parkinson's disease. However, it has barely been investigated in astrocytes. Here, we show by immunofluorescence that both A2A and D2 receptors are expressed in adult rat striatal astrocytes in situ, and investigate on presence, function, and interactions of the receptors in the astrocyte processes-acutely prepared from the adult rat striatum-and on the effects of homocysteine on the A2A-D2 receptor-receptor interaction. We found that A2A and D2 receptors were co-expressed on vesicular glutamate transporter-1-positive astrocyte processes, and confirmed that A2A-D2 receptor-receptor interaction controlled glutamate release-assessed by measuring the [3H]D-aspartate release-from the processes. The complexity of A2A-D2 receptor-receptor interaction is suggested by the effect of intracellular homocysteine, which reduced D2-mediated inhibition of glutamate release (homocysteine allosteric action on D2), without interfering with the A2A-mediated antagonism of the D2 effect (maintained A2A-D2 interaction). Our findings indicate the crucial integrative role of A2A-D2 molecular circuits at the plasma membrane of striatal astrocyte processes. The fact that homocysteine reduced D2-mediated inhibition of glutamate release could provide new insights into striatal astrocyte-neuron intercellular communications. As striatal astrocytes are recognized to be involved in Parkinson's pathophysiology, these findings may shed light on the pathogenic mechanisms of the disease and contribute to the development of new drugs for its treatment.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Padua, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, Section of Biochemistry, and Italian Institute of Biostructures and Biosystems, University of Genova, Genoa, Italy
| | - Carlo Tacchetti
- Experimental Imaging Center, Scientific Institute San Raffaele, Milan, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Susanna Genedani
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Paola Ramoino
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Simone Pelassa
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy. .,Centre of Excellence for Biomedical Research CEBR, University of Genova, Genoa, Italy.
| | - Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Gagnon D, Eid L, Coudé D, Whissel C, Di Paolo T, Parent A, Parent M. Evidence for Sprouting of Dopamine and Serotonin Axons in the Pallidum of Parkinsonian Monkeys. Front Neuroanat 2018; 12:38. [PMID: 29867377 PMCID: PMC5963193 DOI: 10.3389/fnana.2018.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022] Open
Abstract
This light and electron microscopie immunohistochemical quantitative study aimed at determining the state of the dopamine (DA) and serotonin (5-HT) innervations of the internal (GPi) and external (GPe) segments of the pallidum in cynomolgus monkeys (Macaca fascicularis) rendered parkinsonian by systemic injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In contrast to the prominent DA denervation of striatum, the GPi in MPTP monkeys was found to be markedly enriched in DA (TH+) axon varicosities. The posterior sensorimotor region of this major output structure of the basal ganglia was about 8 times more intensely innervated in MPTP monkeys (0.71 ± 0.08 × 106 TH+ axon varicosities/mm3) than in controls (0.09 ± 0.01 × 106). MPTP intoxication also induced a two-fold increase in the density of 5-HT (SERT+) axon varicosities in both GPe and GPi. This augmentation was particularly pronounced anteriorly in the so-called associative and limbic pallidal territories. The total length of the labeled pallidal axons was also significantly increased in MPTP monkeys compared to controls, but the number of DA and 5-HT axon varicosities per axon length unit remained the same in the two groups, indicating that the DA and 5-HT pallidal hyperinnervations seen in MPTP monkeys result from axon sprouting rather than from the appearance of newly formed axon varicosities on non-growing axons. At the ultrastructural level, pallidal TH+ and SERT+ axons were morphologically similar in MPTP and controls, and their synaptic incidence was very low suggesting a volumic mode of transmission. Altogether, our data reveal a significant sprouting of DA and 5-HT pallidal afferents in parkinsonian monkeys, the functional significance of which remains to be determined. We suggest that the marked DA hyperinnervation of the GPi represents a neuroadaptive change designed to normalize pallidal firing patterns associated with the delayed appearance of motor symptoms, whereas the 5-HT hyperinnervation might be involved in the early expression of non-motor symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Dave Gagnon
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Lara Eid
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Dymka Coudé
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Carl Whissel
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Thérèse Di Paolo
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
| | - André Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Martin Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
21
|
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; 365:379-397. [PMID: 29523699 DOI: 10.1124/jpet.117.247171] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/06/2018] [Indexed: 03/08/2025] Open
Abstract
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate is the gold-standard animal model of Parkinson disease (PD) and has been used to assess the effectiveness of experimental drugs on dyskinesia, parkinsonism, and psychosis. Three species have been used in most studies-the macaque, marmoset, and squirrel monkey-the last much less so than the first two species; however, the predictive value of each species at forecasting clinical efficacy, or lack thereof, is poorly documented. Here, we have reviewed all the published literature detailing pharmacologic studies that assessed the effects of experimental drugs on dyskinesia, parkinsonism, and psychosis in each of these species and have calculated their predictive value of success and failure at the clinical level. We found that, for dyskinesia, the macaque has a positive predictive value of 87.5% and a false-positive rate of 38.1%, whereas the marmoset has a positive predictive value of 76.9% and a false-positive rate of 15.6%. For parkinsonism, the macaque has a positive predictive value of 68.2% and a false-positive rate of 44.4%, whereas the marmoset has a positive predictive value of 86.9% and a false-positive rate of 41.7%. No drug that alleviates psychosis in the clinic has shown efficacy at doing so in the macaque, whereas the marmoset has 100% positive predictive value. The small number of studies conducted in the squirrel monkey precluded us from calculating its predictive efficacy. We hope our results will help in the design of pharmacologic experiments and will facilitate the drug discovery and development process in PD.
Collapse
Affiliation(s)
- Nicolas Veyres
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| | - Adjia Hamadjida
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| | - Philippe Huot
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| |
Collapse
|
22
|
Cenci MA, Crossman AR. Animal models of l-dopa-induced dyskinesia in Parkinson's disease. Mov Disord 2018; 33:889-899. [PMID: 29488257 DOI: 10.1002/mds.27337] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/25/2022] Open
Abstract
Understanding the biological mechanisms of l-dopa-induced motor complications is dependent on our ability to investigate these phenomena in animal models of Parkinson's disease. The most common motor complications consist in wearing-off fluctuations and abnormal involuntary movements appearing when plasma levels of l-dopa are high, commonly referred to as peak-dose l-dopa-induced dyskinesia. Parkinsonian models exhibiting these features have been well-characterized in both rodent and nonhuman primate species. The first animal models of peak-dose l-dopa-induced dyskinesia were produced in monkeys lesioned with N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and treated chronically with l-dopa to elicit choreic movements and dystonic postures. Seminal studies were performed in these models using both metabolic mapping and electrophysiological techniques, providing fundamental pathophysiological insights that have stood the test of time. A decade later, it was shown possible to reproduce peak-dose l-dopa-induced dyskinesia in rats and mice rendered parkinsonian with nigrostriatal 6-hydroxydopamine lesions. When treated with l-dopa, these animals exhibit abnormal involuntary movements having both hyperkinetic and dystonic components. These models have enabled molecular- and cellular-level investigations into the mechanisms of l-dopa-induced dyskinesia. A flourishing literature using genetically engineered mice is now unraveling the role of specific genes and neural circuits in the development of l-dopa-induced motor complications. Both non-human primate and rodent models of peak-dose l-dopa-induced dyskinesia have excellent construct validity and provide valuable tools for discovering therapeutic targets and evaluating potential treatments. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- M Angela Cenci
- Department of Experimental Medical Science, Basal Ganglia Pathophysiology Unit, Lund University, Lund, Sweden
| | | |
Collapse
|
23
|
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: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/23/2018] [Indexed: 02/01/2023]
|
24
|
Repetitive transcranial magnetic stimulation (rTMS) improves behavioral and biochemical deficits in levodopa-induced dyskinetic rats model. Oncotarget 2018; 7:58802-58812. [PMID: 27613848 PMCID: PMC5312277 DOI: 10.18632/oncotarget.11587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/21/2016] [Indexed: 11/25/2022] Open
Abstract
Fluctuations of dopamine levels and upregulations of NR2B tyrosine phosphorylation in the striatum have been connected with levodopa (L-dopa)-induced dyskinesia (LID) in Parkinson's disease (PD). Repetitive transcranial magnetic stimulation (rTMS) is one of the noninvasive and potential method treating dyskinesia. Yet, the effect of rTMS on the above key pathological events remains unclear. In this study, we gave L-dopa treatment intraperitoneally for 22 days to 6-hydroxydopamine-lesioned PD rats to prepare LID rats model, and subsequently applied rTMS daily for 3 weeks to LID rats model. The effect of rTMS on abnormal involuntary movements (AIMs) was assessed. After ending the experiments, we further determined tyrosine hydroxylase (TH)-positive dopaminergic neurons number by immunohistochemistry, dopamine levels by HPLC, glial cell line-derived neurotrophic factor (GDNF) levels by ELISA, NR2B tyrosine phosphorylation and interactions of NR2B with Fyn by immunoblotting and immunoprecipitation. The results demonstrated that rTMS obviously attenuated AIMs scores, reduced the loss of nigral dopaminergic neurons and the fluctuations of striatal dopamine levels. Meanwhile, rTMS significantly increased the expression of GDNF, which couldrestore the damage of dopaminergic neurons. Additionally, rTMS also reduced the levels of the NR2B tyrosine phosphorylation andits interactions with Fyn in the lesioned striatum of LID rats model. Thus, these data indicate that rTMS can provide benefit for the therapy of LID by improving the key biochemical deficits related to dyskinesia.
Collapse
|
25
|
Chotibut T, Meadows S, Kasanga E, McInnis T, Cantu MA, Bishop C, Salvatore MF. Ceftriaxone reduces L-dopa-induced dyskinesia severity in 6-hydroxydopamine parkinson's disease model. Mov Disord 2017; 32:1547-1556. [PMID: 28631864 PMCID: PMC5681381 DOI: 10.1002/mds.27077] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Increased extracellular glutamate may contribute to l-dopa induced dyskinesia, a debilitating side effect faced by Parkinson's disease patients 5 to 10 years after l-dopa treatment. Therapeutic strategies targeting postsynaptic glutamate receptors to mitigate dyskinesia may have limited success because of significant side effects. Increasing glutamate uptake may be another approach to attenuate excess glutamatergic neurotransmission to mitigate dyskinesia severity or prolong the time prior to onset. Initiation of a ceftriaxone regimen at the time of nigrostriatal lesion can attenuate tyrosine hydroxylase loss in conjunction with increased glutamate uptake and glutamate transporter GLT-1 expression in a rat 6-hydroxydopamine model. In this article, we examined if a ceftriaxone regimen initiated 1 week after nigrostriatal lesion, but prior to l-dopa, could reduce l-dopa-induced dyskinesia in an established dyskinesia model. METHODS Ceftriaxone (200 mg/kg, intraperitoneal, once daily, 7 consecutive days) was initiated 7 days post-6-hydroxydopamine lesion (days 7-13) and continued every other week (days 21-27, 35-39) until the end of the study (day 39 postlesion, 20 days of l-dopa). RESULTS Ceftriaxone significantly reduced abnormal involuntary movements at 5 time points examined during chronic l-dopa treatment. Partial recovery of motor impairment from nigrostriatal lesion by l-dopa was unaffected by ceftriaxone. The ceftriaxone-treated l-dopa group had significantly increased striatal GLT-1 expression and glutamate uptake. Striatal tyrosine hydroxylase loss in this group was not significantly different when compared with the l-dopa alone group. CONCLUSIONS Initiation of ceftriaxone after nigrostriatal lesion, but prior to and during l-dopa, may reduce dyskinesia severity without affecting l-dopa efficacy or the reduction of striatal tyrosine hydroxylase loss. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tanya Chotibut
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - Samantha Meadows
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000
| | - Ella Kasanga
- Institute for Healthy Aging & Center for Neuroscience Discovery, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Tamara McInnis
- Institute for Healthy Aging & Center for Neuroscience Discovery, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Mark A. Cantu
- Institute for Healthy Aging & Center for Neuroscience Discovery, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000
| | - Michael F. Salvatore
- Institute for Healthy Aging & Center for Neuroscience Discovery, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| |
Collapse
|
26
|
Kong M, Ba M, Ren C, Yu L, Dong S, Yu G, Liang H. An updated meta-analysis of amantadine for treating dyskinesia in Parkinson's disease. Oncotarget 2017; 8:57316-57326. [PMID: 28915672 PMCID: PMC5593643 DOI: 10.18632/oncotarget.17622] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/19/2017] [Indexed: 11/25/2022] Open
Abstract
In recent years, a few of randomized controlled trials (RCTs) about amantadine for treating dyskinesia in Parkinson's disease (PD) were completed. Here, we conducted a systematic literature review about the clinical research to provide the updated evidence for treating dyskinesia. Electronic search of Medline, PubMed, Cochrane Library, and other databases up to May 2016 for relevant studies was performed. We selected the Unified Parkinson's Disease Rating Scale IV (UPDRS IV) and Dyskinesia Rating Scales (DRS) as efficacy outcomes of amantadine on dyskinesia. Pooled data from included studies was then used to carry out meta-analysis. A total of eleven eligible RCTs that involved 356 PD patients with existing dyskinesia were included in the present study. The results of meta-analysis showed that amantadine significantly improved UPDRS IV (P < 0.0001) and DRS (P < 0.00001). Meanwhile, there was a mild reduction in Unified Parkinson's Disease Rating Scale III after amantadine treatment in advanced PD patients with dyskinesia (P = 0.01) compared with placebo. High dosage of amantadine obviously improved existing dyskinesia in PD, yet at the expense of the increased adverse events. It was necessary to detect the optimal therapeutic efficacy to balance the incidence of adverse events when we used amantadine to treat existing dyskinesia in PD patients.
Collapse
Affiliation(s)
- Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| | - Maowen Ba
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong 264000, PR China
| | - Chao Ren
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong 264000, PR China
| | - Ling Yu
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| | - Shengjie Dong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| | - Guoping Yu
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong 264000, PR China
| | - Hui Liang
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| |
Collapse
|
27
|
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: 3.0] [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
|
28
|
Han C, Nie S, Chen G, Ma K, Xiong N, Zhang Z, Xu Y, Wang T, Papa SM, Cao X. Intrastriatal injection of ionomycin profoundly changes motor response to l-DOPA and its underlying molecular mechanisms. Neuroscience 2016; 340:23-33. [PMID: 27771532 DOI: 10.1016/j.neuroscience.2016.10.033] [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] [Received: 09/13/2016] [Revised: 10/08/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Abstract
Long-term l-DOPA treatment of Parkinson's disease is accompanied with fluctuations of motor responses and l-DOPA-induced dyskinesia (LID). Phosphorylation of the dopamine and c-AMP regulated phosphoprotein of 32kDa (DARPP-32) plays a role in the pathogenesis of LID, and thus dephosphorylation of this protein by activated calcineurin may help reduce LID. One important activator of calcineurin is the Ca2+ ionophore ionomycin. Here, we investigated whether intrastriatal injection of ionomycin to hemiparkinsonian rats produced changes in l-DOPA responses including LID. We also analyzed the effects of ionomycin on key molecular mediators of LID. Results confirmed our hypothesis that ionomycin could downregulate the phosphorylation of DARPP32 at Thr-34 and reduce LID. Besides, ionomycin decreased two established molecular markers of LID, FosB/ΔFosB and phosphorylated ERK1/2. Ionomycin also decreased the phosphorylation of three main subunits of the NMDA receptor, NR1 phosphorylated at ser896, NR2A phosphorylated at Tyr-1325, and NR2B phosphorylated at Tyr-1472. Furthermore, the anti-LID effect of striatally injected ionomycin was not accompanied by reduction of the antiparkinsonian action of l-DOPA. These data indicate that ionomycin largely interacts with striatal mechanisms that are critical to the l-DOPA motor response highlighting the role of protein dephosphorylation by calcineurin.
Collapse
Affiliation(s)
- Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuke Nie
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guiqin Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kai Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Stella M Papa
- Yerkes National Primate Research Center, Department of Neurology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
29
|
Petryszyn S, Di Paolo T, Parent A, Parent M. The number of striatal cholinergic interneurons expressing calretinin is increased in parkinsonian monkeys. Neurobiol Dis 2016; 95:46-53. [PMID: 27388937 DOI: 10.1016/j.nbd.2016.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/13/2016] [Accepted: 07/03/2016] [Indexed: 12/17/2022] Open
Abstract
The most abundant interneurons in the primate striatum are those expressing the calcium-binding protein calretinin (CR). The present immunohistochemical study provides detailed assessments of their morphological traits, number, and topographical distribution in normal monkeys (Macaca fascicularis) and in monkeys rendered parkinsonian (PD) by MPTP intoxication. In primates, the CR+ striatal interneurons comprise small (8-12μm), medium (12-20μm) and large-sized (20-45μm) neurons, each with distinctive morphologies. The small CR+ neurons were 2-3 times more abundant than the medium-sized CR+ neurons, which were 20-40 times more numerous than the large CR+ neurons. In normal and PD monkeys, the density of small and medium-sized CR+ neurons was twice as high in the caudate nucleus than in the putamen, whereas the inverse occurred for the large CR+ neurons. Double immunostaining experiments revealed that only the large-sized CR+ neurons expressed choline acetyltransferase (ChAT). The number of large CR+ neurons was found to increase markedly (4-12 times) along the entire anteroposterior extent of both the caudate nucleus and putamen of PD monkeys compared to controls. Comparison of the number of large CR-/ChAT+ and CR+/ChAT+ neurons together with experiments involving the use of bromo-deoxyuridine (BrdU) as a marker of newly generated cells showed that it is the expression of CR by the large ChAT+ striatal interneurons, and not their absolute number, that is increased in the dopamine-depleted striatum. These findings reveal the modulatory role of dopamine in the phenotypic expression of the large cholinergic striatal neurons, which are known to play a crucial role in PD pathophysiology.
Collapse
Affiliation(s)
- Sarah Petryszyn
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - Thérèse Di Paolo
- Centre de recherche du CHU de Québec, Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - André Parent
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - Martin Parent
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
30
|
Benzimidazolone bioisosteres of potent GluN2B selective NMDA receptor antagonists. Eur J Med Chem 2016; 116:136-146. [PMID: 27061977 DOI: 10.1016/j.ejmech.2016.03.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 01/27/2016] [Accepted: 03/21/2016] [Indexed: 11/24/2022]
Abstract
Overactivation of the NMDA receptor is associated with excitotoxic events leading to neurodegenerative processes as observed during the development of Alzheimer's disease, ParFnson's disease, Chorea Huntington and epilepsy. Negative allosteric modulators addressing selectively the ifenprodil binding site of GluN2B subunit containing NMDA receptors are of major interest due to their neuroprotective potential accompanied by few side effects. Herein benzimidazolone bioisosteres of potent GluN2B antagonists 1-5 were designed and synthesized. A seven step sequence provided the central intermediate 19 in 28% yield. Elimination of water, methylation, epoxidation, epoxide rearrangement and finally reductive amination afforded the [7]annulenobenzimidazolone 30 with a 3-phenylpropylamino substituent in 6-position. Although 30 fits nicely into the pharmacophore of potent GluN2B antagonists, the gluN2B binding affinity of 30 was only moderate (Ki = 697 nM). Additionally, 30 shows low selectivity over the σ2 receptor (Ki = 549 nM). The moderate GluN2B affinity was explained by the rigid tricyclic structure of the [7]annulenobenzimidazolone 30.
Collapse
|
31
|
Gagnon D, Gregoire L, Di Paolo T, Parent M. Serotonin hyperinnervation of the striatum with high synaptic incidence in parkinsonian monkeys. Brain Struct Funct 2015; 221:3675-91. [DOI: 10.1007/s00429-015-1125-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/29/2015] [Indexed: 12/23/2022]
|
32
|
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: 359] [Impact Index Per Article: 35.9] [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
|
33
|
Mellone M, Stanic J, Hernandez LF, Iglesias E, Zianni E, Longhi A, Prigent A, Picconi B, Calabresi P, Hirsch EC, Obeso JA, Di Luca M, Gardoni F. NMDA receptor GluN2A/GluN2B subunit ratio as synaptic trait of levodopa-induced dyskinesias: from experimental models to patients. Front Cell Neurosci 2015. [PMID: 26217176 PMCID: PMC4491616 DOI: 10.3389/fncel.2015.00245] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Levodopa-induced dyskinesias (LIDs) are major complications in the pharmacological management of Parkinson's disease (PD). Abnormal glutamatergic transmission in the striatum is considered a key factor in the development of LIDs. This work aims at: (i) characterizing N-methyl-D-aspartate (NMDA) receptor GluN2A/GluN2B subunit ratio as a common synaptic trait in rat and primate models of LIDs as well as in dyskinetic PD patients; and (ii) validating the potential therapeutic effect of a cell-permeable peptide (CPP) interfering with GluN2A synaptic localization on the dyskinetic behavior of these experimental models of LIDs. Here we demonstrate an altered ratio of synaptic GluN2A/GluN2B-containing NMDA receptors in the striatum of levodopa-treated dyskinetic rats and monkeys as well as in post-mortem tissue from dyskinetic PD patients. The modulation of synaptic NMDA receptor composition by a cell-permeable peptide interfering with GluN2A subunit interaction with the scaffolding protein postsynaptic density protein 95 (PSD-95) leads to a reduction in the dyskinetic motor behavior in the two animal models of LIDs. Our results indicate that targeting synaptic NMDA receptor subunit composition may represent an intriguing therapeutic approach aimed at ameliorating levodopa motor side effects.
Collapse
Affiliation(s)
- Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| | - Jennifer Stanic
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| | - Ledia F Hernandez
- Movement Disorders Group, Neurosciences Division, Center for Applied Medical Research (CIMA), Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), University of Navarra Pamplona, Spain
| | - Elena Iglesias
- Movement Disorders Group, Neurosciences Division, Center for Applied Medical Research (CIMA), Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), University of Navarra Pamplona, Spain
| | - Elisa Zianni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| | - Annalisa Longhi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| | - Annick Prigent
- Inserm, U 1127 Paris, France ; CNRS, UMR 7225 Paris, France ; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127 Paris, France ; Institut du Cerveau et de la Moelle Épinière, ICM Paris, France
| | | | - Paolo Calabresi
- Fondazione Santa Lucia, IRCCS Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, Localitá, Sant'Andrea delle Fratte Perugia, Italy
| | - Etienne C Hirsch
- Inserm, U 1127 Paris, France ; CNRS, UMR 7225 Paris, France ; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127 Paris, France ; Institut du Cerveau et de la Moelle Épinière, ICM Paris, France
| | - Jose A Obeso
- Movement Disorders Group, Neurosciences Division, Center for Applied Medical Research (CIMA), Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), University of Navarra Pamplona, Spain
| | - Monica Di Luca
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano Milano, Italy
| |
Collapse
|
34
|
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: 5] [Impact Index Per Article: 0.5] [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
|
35
|
Kong M, Ba M, Liu C, Zhang Y, Zhang H, Qiu H. NR2B antagonist CP-101,606 inhibits NR2B phosphorylation at tyrosine-1472 and its interactions with Fyn in levodopa-induced dyskinesia rat model. Behav Brain Res 2015; 282:46-53. [PMID: 25576965 DOI: 10.1016/j.bbr.2014.12.059] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/28/2014] [Accepted: 12/31/2014] [Indexed: 10/24/2022]
Abstract
The augmented tyrosine phosphorylation of NR2B subunit of N-methyl-d-aspartate receptors (NMDAR) dependent on Fyn kinase has been associated with levodopa (l-dopa)-induced dyskinesia (LID). CP-101,606, one selective NR2B subunit antagonist, can improve dyskinesia. Yet, the accurate action mechanism is less well understood. In the present study, the evidences were investigated. Valid 6-hydroxydopamine-lesioned parkinsonian rats were treated with l-dopa intraperitoneally for 22 days to induce LID rat model. On day 23, rats received either CP-101,606 (0.5mg/kg) or vehicle with each l-dopa dose. On the day of 1, 8, 15, 22, and 23 during l-dopa treatment, we determined abnormal involuntary movements (AIMs) in rats. The levels of NR2B phosphorylation at tyrosine-1472 (pNR2B-Tyr1472) and interactions of NR2B with Fyn in LID rat model were detected by immunoblotting and immunoprecipitation. Results showed that CP-101,606 attenuated l-dopa-induced AIMs. In agreement with behavioral analysis, CP-101,606 reduced the augmented pNR2B-Tyr1472 and its interactions with Fyn triggered during the l-dopa administration in the lesioned striatum of parkinsonian rats. Moreover, CP-101,606 also decreased the level of Ca(2+)/calmodulin-dependent protein kinase II at threonine-286 hyperphosphorylation (pCaMKII-Thr286), which was the downstream signaling amplification molecule of NMDAR overactivation and closely associated with LID. However, the protein level of NR2B and Fyn had no difference under the above conditions. These data indicate that the inhibition of the interactions of NR2B with Fyn and NR2B tyrosine phosphorylation may contribute to the CP-101,606-induced downregulation of NMDAR function and provide benefit for the therapy of LID.
Collapse
Affiliation(s)
- Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, China
| | - Maowen Ba
- Department of Neurology, Yuhuangding Hospital, Yantai City, Shandong 264000, China.
| | - Chuanyu Liu
- Department of Neurology, Yuhuangding Hospital, Yantai City, Shandong 264000, China
| | - Yanxiang Zhang
- Department of Neurology, Yuhuangding Hospital, Yantai City, Shandong 264000, China
| | - Hongli Zhang
- Department of Endocrinology, Ruijin Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Haiyan Qiu
- Department of Neuroscience, the University of Texas Southwestern Medical Center, TX 75390, USA
| |
Collapse
|
36
|
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
|
37
|
Gardoni F, Di Luca M. Targeting glutamatergic synapses in Parkinson's disease. Curr Opin Pharmacol 2014; 20:24-8. [PMID: 25462288 DOI: 10.1016/j.coph.2014.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons of the substantia nigra and dramatic motor and cognitive impairments. The current knowledge indicates that the strength of glutamatergic signals from the cortex to the striatum is regulated during the progression of the disease. The efficacy of ionotropic glutamate receptors to modulate synaptic transmission in the striatum indicates that modulation of the activity of these receptors may represent a key target to rescue the altered neurotransmission in PD. Preclinical and clinical studies suggest that agents targeting ionotropic glutamate receptors may ameliorate the motor symptoms of PD as well as to reduce the onset of levodopa-induced dyskinetic motor behaviour.
Collapse
Affiliation(s)
- Fabrizio Gardoni
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Monica Di Luca
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
38
|
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: 2.3] [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
|
39
|
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: 6.1] [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]
|
40
|
Majláth Z, Vécsei L. NMDA antagonists as Parkinson’s disease therapy: disseminating the evidence. Neurodegener Dis Manag 2014; 4:23-30. [DOI: 10.2217/nmt.13.77] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Oral levodopa is the current baseline therapy in the management of Parkinson’s disease, but nonmotor complications and therapy-related dyskinesias pose an important challenge for clinicians. Glutamate receptors have been implicated in the neurodegenerative process of Parkinson’s disease and also in the development of levodopa-induced dyskinesias. This article discusses the role of NMDA receptors in Parkinson’s disease and their modulation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Zsófia Majláth
- Department of Neurology, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - László Vécsei
- Neuroscience Research Group of the Hungarian Academy of Sciences & University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| |
Collapse
|
41
|
Koutsokera M, Kafkalias P, Giompres P, Kouvelas ED, Mitsacos A. Expression and phosphorylation of glutamate receptor subunits and CaMKII in a mouse model of Parkinsonism. Brain Res 2014; 1549:22-31. [PMID: 24418465 DOI: 10.1016/j.brainres.2013.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/24/2013] [Accepted: 12/19/2013] [Indexed: 12/29/2022]
Abstract
Dopaminergic deficiency of the weaver mutant mouse is a valuable tool to further our understanding of Parkinson׳s disease (PD) pathogenesis since dopaminergic neurons of the nigrostriatal pathway undergo spontaneous and progressive cell death. In the present study we investigated the changes in protein expression and phosphorylation of glutamate receptor subunits and αCaMKII in weaver striatum at the end of the third and sixth postnatal month. Using immunoblotting, we found increased immunoreactivity levels of both GluN2A and GluN2B subunits of NMDA receptors and GluA1 subunit of AMPA receptors approximately from 75% to 110% in the 3-month-old weaver striatum compared to control. In the 6-month-old weaver striatum, no changes were detected in GluN2A and GluA1 immunoreactivity levels, whereas GluN2B showed a 21% statistically significant increase. Our results also indicated increased phospho-S1303 GluN2B in both 3 and 6 month-olds and increased phospho-S831 and -845 GluA1 in 3 month-old weaver striatum. However, these increases did not exceed the increases observed for total GluN2B and GluA1. Furthermore, our results showed increased immunoreactivity levels for phospho-T286 αCaMKII by approximately 180% in the 6 month-old weaver striatum, while total CaMKII immunoreactivity levels were not altered at either 3- or 6-month-old weaver. Our results suggest that distinct degrees of DA neuron degeneration differentially affect expression and phosphorylation of striatal glutamate receptors and αCaMKII. Findings on this genetic parkinsonian model suggest that striatal glutamatergic signaling may play an important role in synaptic plasticity and motor behavior that follow progressive and chronic dopamine depletion in PD with biochemical consequences beyond those seen in acute toxic models.
Collapse
Affiliation(s)
- Maria Koutsokera
- Laboratory of Physiology, Faculty of Medicine, University of Patras, 26500 Patras, Greece
| | - Panagiotis Kafkalias
- Laboratory of Physiology, Faculty of Medicine, University of Patras, 26500 Patras, Greece
| | - Panagiotis Giompres
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, 26500 Patras, Greece
| | - Elias D Kouvelas
- Laboratory of Physiology, Faculty of Medicine, University of Patras, 26500 Patras, Greece
| | - Ada Mitsacos
- Laboratory of Physiology, Faculty of Medicine, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
42
|
Carrillo-Mora P, Silva-Adaya D, Villaseñor-Aguayo K. Glutamate in Parkinson's disease: Role of antiglutamatergic drugs. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.baga.2013.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
43
|
Performance of movement in hemiparkinsonian rats influences the modifications induced by dopamine agonists in striatal efferent dynorphinergic neurons. Exp Neurol 2013; 247:663-72. [DOI: 10.1016/j.expneurol.2013.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/20/2013] [Accepted: 03/02/2013] [Indexed: 11/22/2022]
|
44
|
Porras G, Li Q, Bezard E. Modeling Parkinson's disease in primates: The MPTP model. Cold Spring Harb Perspect Med 2013; 2:a009308. [PMID: 22393538 DOI: 10.1101/cshperspect.a009308] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) primate models of Parkinson's disease (PD) reproduce most, although not all, of the clinical and pathological hallmarks of PD. The present contribution presents the possibilities offered by the MPTP monkey models of PD to readers with minimal knowledge of PD, emphasizing the diversity of species, route and regimen of administration, symptoms and pathological features. Readers would eventually find out that there is not a single MPTP monkey model of PD but instead MPTP monkey models of PD, each addressing a specific experimental need.
Collapse
Affiliation(s)
- Gregory Porras
- University de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France; CNRS, Institut des Maladiesw Neurodégénératives, UMR 5293 Bordeaux, France
| | | | | |
Collapse
|
45
|
Mellone M, Gardoni F. Modulation of NMDA receptor at the synapse: promising therapeutic interventions in disorders of the nervous system. Eur J Pharmacol 2013; 719:75-83. [PMID: 23872417 DOI: 10.1016/j.ejphar.2013.04.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 03/20/2013] [Accepted: 04/03/2013] [Indexed: 12/25/2022]
Abstract
There is general agreement that excessive activation of N-methyl-D-aspartate (NMDA) receptors plays a key role in mediating at least some aspects of synaptic dysfunction in several central nervous system disorders. On this view, in the last decades, research focused on the discovery of different compounds able to reduce NMDA receptor activity, such as classical and/or subunit-specific antagonists. However, the increasing body of knowledge on specific signaling pathways downstream NMDA receptors led to the identification of new pharmacological targets for NMDA receptor-related pathological conditions. Moreover, besides over-activation, several studies indicated that also abnormal NMDA receptor trafficking, resulting in the modification of the receptor subunit composition at the synapse, has a major role in the pathogenesis of several brain disorders. For this reason, the discovery of the molecular mechanisms regulating the abundance of synaptic versus extra-synaptic NMDA receptors as well as the activation of the specific signaling pathways downstream the different NMDA receptor subtypes is needed for the development of novel therapeutic approaches for NMDA receptor-dependent synaptic dysfunction.
Collapse
Affiliation(s)
- Manuela Mellone
- Dipartimento Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milano, Italy.
| |
Collapse
|
46
|
Gasparini F, Di Paolo T, Gomez-Mancilla B. Metabotropic glutamate receptors for Parkinson's disease therapy. PARKINSON'S DISEASE 2013; 2013:196028. [PMID: 23853735 PMCID: PMC3703788 DOI: 10.1155/2013/196028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/29/2013] [Indexed: 12/21/2022]
Abstract
Excessive glutamatergic signalling within the basal ganglia is implicated in the progression of Parkinson's disease (PD) and inthe emergence of dyskinesia associated with long-term treatment with L-DOPA. There is considerable research focus on the discovery and development of compounds that modulate glutamatergic signalling via glutamate receptors, as treatments for PD and L-DOPA-induced dyskinesia (LID). Although initial preclinical studies with ionotropic glutamate receptor antagonists showed antiparkinsonian and antidyskinetic activity, their clinical use was limited due to psychiatric adverse effects, with the exception of amantadine, a weak N-methyl-d-aspartate (NMDA) antagonist, currently used to reduce dyskinesia in PD patients. Metabotropic receptor (mGlu receptor) modulators were considered to have a more favourable side-effect profile, and several agents have been studied in preclinical models of PD. The most promising results have been seen clinically with selective antagonists of mGlu5 receptor and preclinically with selective positive allosteric modulators of mGlu4 receptor. The growing understanding of glutamate receptor crosstalk also raises the possibility of more precise modulation of glutamatergic transmission, which may lead to the development of more effective agents for PD.
Collapse
Affiliation(s)
- Fabrizio Gasparini
- Novartis Pharma AG, Novartis Institutes for BioMedical Research Basel, Forum 1, Novartis Campus, 4056 Basel, Switzerland
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, QC, Canada G1V 4G2
- Faculty of Pharmacy, Laval University, Quebec City, QC, Canada G1K 7P4
| | - Baltazar Gomez-Mancilla
- Novartis Pharma AG, Novartis Institutes for BioMedical Research Basel, Forum 1, Novartis Campus, 4056 Basel, Switzerland
| |
Collapse
|
47
|
Wu N, Song L, Yang X, Yuan W, Liu Z. NMDA receptor regulation of levodopa-induced behavior and changes in striatal G protein-coupled receptor kinase 6 and β-arrestin-1 expression in parkinsonian rats. Clin Interv Aging 2013; 8:347-52. [PMID: 23569367 PMCID: PMC3615843 DOI: 10.2147/cia.s41464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Parkinson's disease is a neurodegenerative disorder caused by loss of dopaminergic neurons in the substantia nigra. The dopamine precursor, levodopa, remains the most effective and common treatment for this disorder. However, long-term administration of levodopa is known to induce characteristic dyskinesia, and molecular mechanisms underlying dyskinesia are poorly understood. METHODS In this study, we investigated the effect of 6-hydroxydopamine lesions in dopaminergic neurons and chronic treatment with levodopa on expression of G protein-coupled receptor kinase 6 and β-arrestin-1, two key regulators of G protein-coupled receptors, in the rat striatum. RESULTS We found that a unilateral 6-hydroxydopamine lesion reduced expression of G protein-coupled receptor kinase 6 and β-arrestin-1 protein in the lesioned striatum. Reduction of these two proteins persisted in 6-hydroxydopamine-lesioned rats on chronic levodopa treatment for 23 days. In addition, coadministration of the N-methyl-D-aspartate receptor antagonist, MK-801, and levodopa reversed the reduction of G protein-coupled receptor kinase 6 and β-arrestin-1 in the striatum. MK-801 also attenuated levodopa-induced dyskinetic behavior. CONCLUSION These data indicate that G protein-coupled receptor kinase 6 and β-arrestin-1 in striatal neurons are sensitive to dopamine depletion and are downregulated in rats with Parkinson's disease and in levodopa-treated rats with the disease. This downregulation seems to require activation of N-methyl-D-aspartate glutamate receptors.
Collapse
Affiliation(s)
- Na Wu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
48
|
Nevalainen N, Lundblad M, Gerhardt GA, Strömberg I. Striatal glutamate release in L-DOPA-induced dyskinetic animals. PLoS One 2013; 8:e55706. [PMID: 23390548 PMCID: PMC3563586 DOI: 10.1371/journal.pone.0055706] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 12/28/2012] [Indexed: 11/18/2022] Open
Abstract
L-DOPA-induced dyskinesia is a common side effect developed after chronic treatment with 3,4-dihydroxyphenyl-l-alanine (l-DOPA) in Parkinson's disease. The biological mechanisms behind this side effect are not fully comprehended although involvement of dopaminergic, serotonergic, and glutamatergic systems has been suggested. The present study utilizes in vivo amperometry to investigate the impact from unilateral 6-hydroxydopamine lesions and l-DOPA (4 mg/kg, including benserazide 15 mg/kg) -induced dyskinetic behavior on striatal basal extracellular glutamate concentration and potassium-evoked glutamate release in urethane-anesthetized rats. Recordings were performed before and after local L-DOPA application in the striatum. In addition, effects from the 5-HT(1A) receptor agonist (2R)-(+)-8-hydroxy-2-(di-n-propylamino)tetralin hydrobromide (8-OHDPAT; 1 mg/kg) was assessed on glutamate release and on dyskinetic behavior. The results revealed a bilateral ≈ 30% reduction of basal extracellular glutamate concentration and attenuated potassium-evoked glutamate release after a unilateral dopamine-depletion in L-DOPA naïve animals. In dyskinetic subjects, basal glutamate concentration was comparable to normal controls, although potassium-evoked glutamate release was reduced to similar levels as in drug naïve dopamine-lesioned animals. Furthermore, acute striatal L-DOPA administration attenuated glutamate release in all groups, except in the dopamine-lesioned striatum of dyskinetic animals. Co-administration of 8-OHDPAT and L-DOPA decreased dyskinesia in dopamine-lesioned animals, but did not affect potassium-evoked glutamate release, which was seen in normal animals. These findings indicate altered glutamate transmission upon dopamine-depletion and dyskinesia.
Collapse
Affiliation(s)
| | - Martin Lundblad
- Basal Ganglia Pathophysiology Unit, Neuroscience Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Greg A. Gerhardt
- Anatomy, Neurobiology, and Neurology, University of Kentucky Medical Center, Lexington, Kentucky, United States of America
| | - Ingrid Strömberg
- Integrative Medical Biology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
49
|
Huot P, Johnston TH, Koprich JB, Fox SH, Brotchie JM. The pharmacology of L-DOPA-induced dyskinesia in Parkinson's disease. Pharmacol Rev 2013; 65:171-222. [PMID: 23319549 DOI: 10.1124/pr.111.005678] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
L-3,4-Dihydroxyphenylalanine (L-DOPA) remains the most effective symptomatic treatment of Parkinson's disease (PD). However, long-term administration of L-DOPA is marred by the emergence of abnormal involuntary movements, i.e., L-DOPA-induced dyskinesia (LID). Years of intensive research have yielded significant progress in the quest to elucidate the mechanisms leading to the development and expression of dyskinesia and maintenance of the dyskinetic state, but the search for a complete understanding is still ongoing. Herein, we summarize the current knowledge of the pharmacology of LID in PD. Specifically, we review evidence gathered from postmortem and pharmacological studies, both preclinical and clinical, and discuss the involvement of dopaminergic and nondopaminergic systems, including glutamatergic, opioid, serotonergic, γ-aminobutyric acid (GABA)-ergic, adenosine, cannabinoid, adrenergic, histaminergic, and cholinergic systems. Moreover, we discuss changes occurring in transcription factors, intracellular signaling, and gene expression in the dyskinetic phenotype. Inasmuch as a multitude of neurotransmitters and receptors play a role in the etiology of dyskinesia, we propose that to optimally alleviate this motor complication, it may be necessary to develop combined treatment approaches that will target simultaneously more than one neurotransmitter system. This could be achieved via three ways as follows: 1) by developing compounds that will interact simultaneously to a multitude of receptors with the required agonist/antagonist effect at each target, 2) by targeting intracellular signaling cascades where the signals mediated by multiple receptors converge, and/or 3) to regulate gene expression in a manner that has effects on signaling by multiple pathways.
Collapse
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
50
|
Effects of L-DOPA and STN-HFS dyskinesiogenic treatments on NR2B regulation in basal ganglia in the rat model of Parkinson's disease. Neurobiol Dis 2012; 48:379-90. [DOI: 10.1016/j.nbd.2012.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/06/2012] [Accepted: 06/22/2012] [Indexed: 11/22/2022] Open
|