1
|
di Biase L, Pecoraro PM, Carbone SP, Caminiti ML, Di Lazzaro V. Levodopa-Induced Dyskinesias in Parkinson's Disease: An Overview on Pathophysiology, Clinical Manifestations, Therapy Management Strategies and Future Directions. J Clin Med 2023; 12:4427. [PMID: 37445461 DOI: 10.3390/jcm12134427] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/18/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Since its first introduction, levodopa has become the cornerstone for the treatment of Parkinson's disease and remains the leading therapeutic choice for motor control therapy so far. Unfortunately, the subsequent appearance of abnormal involuntary movements, known as dyskinesias, is a frequent drawback. Despite the deep knowledge of this complication, in terms of clinical phenomenology and the temporal relationship during a levodopa regimen, less is clear about the pathophysiological mechanisms underpinning it. As the disease progresses, specific oscillatory activities of both motor cortical and basal ganglia neurons and variation in levodopa metabolism, in terms of the dopamine receptor stimulation pattern and turnover rate, underlie dyskinesia onset. This review aims to provide a global overview on levodopa-induced dyskinesias, focusing on pathophysiology, clinical manifestations, therapy management strategies and future directions.
Collapse
Affiliation(s)
- Lazzaro di Biase
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Brain Innovations Lab, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Pasquale Maria Pecoraro
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Simona Paola Carbone
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Maria Letizia Caminiti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Vincenzo Di Lazzaro
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
2
|
Kwan C, Kang W, Kim E, Belliveau S, Frouni I, Huot P. Metabotropic glutamate receptors in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:1-31. [PMID: 36868628 DOI: 10.1016/bs.irn.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Parkinson's disease (PD) is a complex disorder that leads to alterations in multiple neurotransmitter systems, notably glutamate. As such, several drugs acting at glutamatergic receptors have been assessed to alleviate the manifestation of PD and treatment-related complications, culminating with the approval of the N-methyl-d-aspartate (NMDA) antagonist amantadine for l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia. Glutamate elicits its actions through several ionotropic and metabotropic (mGlu) receptors. There are 8 sub-types of mGlu receptors, with sub-types 4 (mGlu4) and 5 (mGlu5) modulators having been tested in the clinic for endpoints pertaining to PD, while sub-types 2 (mGlu2) and 3 (mGlu3) have been investigated in pre-clinical settings. In this book chapter, we provide an overview of mGlu receptors in PD, with a focus on mGlu5, mGlu4, mGlu2 and mGlu3 receptors. For each sub-type, we review, when applicable, their anatomical localization and possible mechanisms underlying their efficacy for specific disease manifestation or treatment-induced complications. We then summarize the findings of pre-clinical studies and clinical trials with pharmacological agents and discuss the potential strengths and limitations of each target. We conclude by offering some perspectives on the potential use of mGlu modulators in the treatment of PD.
Collapse
Affiliation(s)
- Cynthia Kwan
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Woojin Kang
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Esther Kim
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Sébastien Belliveau
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Movement Disorder Clinic, Division of Neurology, Department of Neurosciences, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
3
|
Alosaimi F, Boonstra JT, Tan S, Temel Y, Jahanshahi A. The role of neurotransmitter systems in mediating deep brain stimulation effects in Parkinson’s disease. Front Neurosci 2022; 16:998932. [PMID: 36278000 PMCID: PMC9579467 DOI: 10.3389/fnins.2022.998932] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Deep brain stimulation (DBS) is among the most successful paradigms in both translational and reverse translational neuroscience. DBS has developed into a standard treatment for movement disorders such as Parkinson’s disease (PD) in recent decades, however, specific mechanisms behind DBS’s efficacy and side effects remain unrevealed. Several hypotheses have been proposed, including neuronal firing rate and pattern theories that emphasize the impact of DBS on local circuitry but detail distant electrophysiological readouts to a lesser extent. Furthermore, ample preclinical and clinical evidence indicates that DBS influences neurotransmitter dynamics in PD, particularly the effects of subthalamic nucleus (STN) DBS on striatal dopaminergic and glutamatergic systems; pallidum DBS on striatal dopaminergic and GABAergic systems; pedunculopontine nucleus DBS on cholinergic systems; and STN-DBS on locus coeruleus (LC) noradrenergic system. DBS has additionally been associated with mood-related side effects within brainstem serotoninergic systems in response to STN-DBS. Still, addressing the mechanisms of DBS on neurotransmitters’ dynamics is commonly overlooked due to its practical difficulties in monitoring real-time changes in remote areas. Given that electrical stimulation alters neurotransmitter release in local and remote regions, it eventually exhibits changes in specific neuronal functions. Consequently, such changes lead to further modulation, synthesis, and release of neurotransmitters. This narrative review discusses the main neurotransmitter dynamics in PD and their role in mediating DBS effects from preclinical and clinical data.
Collapse
Affiliation(s)
- Faisal Alosaimi
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
- *Correspondence: Faisal Alosaimi,
| | - Jackson Tyler Boonstra
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Sonny Tan
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Ali Jahanshahi
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Ali Jahanshahi,
| |
Collapse
|
4
|
Garin CM, Nadkarni NA, Pépin J, Flament J, Dhenain M. Whole brain mapping of glutamate distribution in adult and old primates at 11.7T. Neuroimage 2022; 251:118984. [PMID: 35149230 DOI: 10.1016/j.neuroimage.2022.118984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
Glutamate is the amino acid with the highest cerebral concentration. It plays a central role in brain metabolism. It is also the principal excitatory neurotransmitter in the brain and is involved in multiple cognitive functions. Alterations of the glutamatergic system may contribute to the pathophysiology of many neurological disorders. For example, changes of glutamate availability are reported in rodents and humans during Alzheimer's and Huntington's diseases, epilepsy as well as during aging. Most studies evaluating cerebral glutamate have used invasive or spectroscopy approaches focusing on specific brain areas. Chemical Exchange Saturation Transfer imaging of glutamate (gluCEST) is a recently developed imaging technique that can be used to study relative changes in glutamate distribution in the entire brain with higher sensitivity and at higher resolution than previous techniques. It thus has strong potential clinical applications to assess glutamate changes in the brain. High field is a key condition to perform gluCEST images with a meaningful signal to noise ratio. Thus, even if some studies started to evaluate gluCEST in humans, most studies focused on rodent models that can be imaged at high magnetic field. In particular, systematic characterization of gluCEST contrast distribution throughout the whole brain has never been performed in humans or non-human primates. Here, we characterized for the first time the distribution of the gluCEST contrast in the whole brain and in large-scale networks of mouse lemur primates at 11.7 Tesla. Because of its small size, this primate can be imaged in high magnetic field systems. It is widely studied as a model of cerebral aging or Alzheimer's disease. We observed high gluCEST contrast in cerebral regions such as the nucleus accumbens, septum, basal forebrain, cortical areas 24 and 25. Age-related alterations of this biomarker were detected in the nucleus accumbens, septum, basal forebrain, globus pallidus, hypophysis, cortical areas 24, 21, 6 and in olfactory bulbs. An age-related gluCEST contrast decrease was also detected in specific neuronal networks, such as fronto-temporal and evaluative limbic networks. These results outline regional differences of gluCEST contrast and strengthen its potential to provide new biomarkers of cerebral function in primates.
Collapse
Affiliation(s)
- Clément M Garin
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France
| | - Nachiket A Nadkarni
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France
| | - Jérémy Pépin
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France
| | - Julien Flament
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France
| | - Marc Dhenain
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, F-92265 Fontenay-aux-Roses, France.
| |
Collapse
|
5
|
Guerra A, Asci F, Zampogna A, D'Onofrio V, Suppa A, Fabbrini G, Berardelli A. Long-term changes in short-interval intracortical facilitation modulate motor cortex plasticity and L-dopa-induced dyskinesia in Parkinson's disease. Brain Stimul 2021; 15:99-108. [PMID: 34823038 DOI: 10.1016/j.brs.2021.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Abnormal glutamatergic neurotransmission in the primary motor cortex (M1) contributes to Parkinson's disease (PD) pathophysiology and is related to l-dopa-induced dyskinesia (LID). We previously showed that short-term treatment with safinamide, a monoamine oxidase type-B inhibitor with anti-glutamatergic properties, improves abnormally enhanced short-interval intracortical facilitation (SICF) in PD patients. OBJECTIVE To examine whether a long-term SICF modulation has beneficial effects on clinical measures, including LID severity, and whether these changes parallel improvement in cortical plasticity mechanisms in PD. METHODS We tested SICF in patients with and without LID before (S0) and after short- (14 days - S1) and long-term (12 months - S2) treatment with safinamide 100 mg/day. Possible changes in M1 plasticity were assessed using intermittent theta-burst stimulation (iTBS). Finally, we correlated safinamide-related neurophysiological changes with modifications in clinical scores. RESULTS SICF was enhanced at S0, and prominently in patients with LID. Safinamide normalized SICF at S1, and this effect persisted at S2. Impaired iTBS-induced plasticity was present at S0 and safinamide restored this alteration at S2. There was a significant correlation between the degree of SICF and the amount of iTBS-induced plasticity at S0 and S2. In patients with LID, the degree of SICF at S0 and S2 correlated with long-term changes in LID severity. CONCLUSIONS Altered SICF contributes to M1 plasticity impairment in PD. Both SICF and M1 plasticity improve after long-term treatment with safinamide. The abnormality in SICF-related glutamatergic circuits plays a role in LID pathophysiology, and its long-term modulation may prevent LID worsening over time.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Suppa
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Human Neurosciences, Sapienza University of Rome, Italy
| | - Giovanni Fabbrini
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Human Neurosciences, Sapienza University of Rome, Italy
| | - Alfredo Berardelli
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Human Neurosciences, Sapienza University of Rome, Italy.
| |
Collapse
|
6
|
Boccella S, Marabese I, Guida F, Luongo L, Maione S, Palazzo E. The Modulation of Pain by Metabotropic Glutamate Receptors 7 and 8 in the Dorsal Striatum. Curr Neuropharmacol 2020; 18:34-50. [PMID: 31210112 PMCID: PMC7327935 DOI: 10.2174/1570159x17666190618121859] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 05/31/2019] [Indexed: 12/28/2022] Open
Abstract
The dorsal striatum, apart from controlling voluntary movement, displays a recently demonstrated pain inhibition. It is connected to the descending pain modulatory system and in particular to the rostral ventromedial medulla through the medullary dorsal reticular nucleus. Diseases of the basal ganglia, such as Parkinson's disease, in addition to being characterized by motor disorders, are associated with pain and hyperactivation of the excitatory transmission. A way to counteract glutamatergic hyperactivation is through the activation of group III metabotropic glutamate receptors (mGluRs), which are located on presynaptic terminals inhibiting neurotransmitter release. So far the mGluRs of group III have been the least investigated, owing to a lack of selective tools. More recently, selective ligands for each mGluR of group III, in particular positive and negative allosteric modulators, have been developed and the role of each subtype is starting to emerge. The neuroprotective potential of group III mGluRs in pathological conditions, such as those characterized by elevate glutamate, has been recently shown. In the dorsal striatum, mGluR7 and mGluR8 are located at glutamatergic corticostriatal terminals and their stimulation inhibits pain in pathological conditions such as neuropathic pain. The two receptors in the dorsal striatum have instead a different role in pain control in normal conditions. This review will discuss recent results focusing on the contribution of mGluR7 and mGluR8 in the dorsal striatal control of pain. The role of mGluR4, whose antiparkinsonian activity is widely reported, will also be addressed.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
7
|
Guerra A, Suppa A, D'Onofrio V, Di Stasio F, Asci F, Fabbrini G, Berardelli A. Abnormal cortical facilitation and L-dopa-induced dyskinesia in Parkinson's disease. Brain Stimul 2019; 12:1517-1525. [DOI: 10.1016/j.brs.2019.06.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/05/2019] [Accepted: 06/07/2019] [Indexed: 10/26/2022] Open
|
8
|
Metabotropic Glutamate Receptor 5 and 8 Modulate the Ameliorative Effect of Ultramicronized Palmitoylethanolamide on Cognitive Decline Associated with Neuropathic Pain. Int J Mol Sci 2019; 20:ijms20071757. [PMID: 30970677 PMCID: PMC6480075 DOI: 10.3390/ijms20071757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/23/2022] Open
Abstract
This study investigated whether metabotropic glutamate receptor (mGluR) 5 and 8 are involved in the effect of ultramicronizedpalmitoylethanolamide (um-PEA) on the cognitive behavior and long term potentiation (LTP) at entorhinal cortex (LEC)-dentate gyrus (DG) pathway in mice rendered neuropathic by the spare nerve injury (SNI). SNI reduced discriminative memory and LTP. Um-PEA treatment started after the development of neuropathic pain had no effects in sham mice, whereas it restored cognitive behavior and LTP in SNI mice. 2-Methyl-6-(phenylethynyl) pyridine (MPEP), a selective mGluR5 antagonist, improved cognition in SNI mice and produced a chemical long term depression of the field excitatory postsynaptic potentials (fEPSPs) in sham and SNI mice. After theta burst stimulation (TBS) MPEP restored LTP in SNI mice. In combination with PEA, MPEP antagonized the PEA effect on discriminative memory and decreased LTP in SNI mice. The (RS)-4-(1-amino-1-carboxyethyl)phthalic acid (MDCPG), a selective mGluR8 antagonist, did not affect discriminative memory, but it induced a chemical LTP and prevented the enhancement of fEPSPs after TBS in SNI mice which were treated or not treated with PEA. The effect of PEA on LTP and cognitive behavior was modulated by mGluR5 and mGluR8. In particular in the SNI conditions, the mGluR5 blockade facilitated memory and LTP, but prevented the beneficial effects of PEA on discriminative memory while the mGluR8 blockade, which was ineffective in itself, prevented the favorable action of the PEA on LTP. Thus, although their opposite roles (excitatory/inhibitory of the two receptor subtypes on the glutamatergic system), they appeared to be required for the neuroprotective effect of PEA in conditions of neuropathic pain.
Collapse
|
9
|
Godlewska BR, Masaki C, Sharpley AL, Cowen PJ, Emir UE. Brain glutamate in medication-free depressed patients: a proton MRS study at 7 Tesla. Psychol Med 2018; 48:1731-1737. [PMID: 29224573 PMCID: PMC6088784 DOI: 10.1017/s0033291717003373] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The possible role of glutamate in the pathophysiology and treatment of depression is of intense current interest. Proton magnetic resonance spectroscopy (MRS) enables the detection of glutamate in the living human brain and meta-analyses of previous MRS studies in depressed patients have suggested that glutamate levels are decreased in anterior brain regions. Nevertheless, at conventional magnetic field strengths [1.5-3 Tesla (T)], it is difficult to separate glutamate from its metabolite and precursor, glutamine, with the two often being measured together as Glx. In contrast, MRS at 7 T allows clear spectral resolution of glutamate and glutamine. METHOD We studied 55 un-medicated depressed patients and 50 healthy controls who underwent MRS scanning at 7 T with voxels placed in anterior cingulate cortex, occipital cortex and putamen (PUT). Neurometabolites were calculated using the unsuppressed water signal as a reference. RESULTS Compared with controls, depressed patients showed no significant difference in glutamate in any of the three voxels studied; however, glutamine concentrations in the patients were elevated by about 12% in the PUT (p < 0.001). CONCLUSIONS The increase in glutamine in PUT is of interest in view of the postulated role of the basal ganglia in the neuropsychology of depression and is consistent with elevated activity in the descending cortical glutamatergic innervation to the PUT. The basal ganglia have rarely been the subject of MRS investigations in depressed patients and further MRS studies of these structures in depression are warranted.
Collapse
Affiliation(s)
- Beata R. Godlewska
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Charles Masaki
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Ann L. Sharpley
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Philip J. Cowen
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Uzay E. Emir
- Oxford Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
10
|
Picconi B, Hernández LF, Obeso JA, Calabresi P. Motor complications in Parkinson's disease: Striatal molecular and electrophysiological mechanisms of dyskinesias. Mov Disord 2017; 33:867-876. [PMID: 29219207 DOI: 10.1002/mds.27261] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022] Open
Abstract
Long-term levodopa (l-dopa) treatment in patients with Parkinson´s disease (PD) is associated with the development of motor complications (ie, motor fluctuations and dyskinesias). The principal etiopathogenic factors are the degree of nigro-striatal dopaminergic loss and the duration and dose of l-dopa treatment. In this review article we concentrate on analysis of the mechanisms underlying l-dopa-induced dyskinesias, a phenomenon that causes disability in a proportion of patients and that has not benefited from major therapeutic advances. Thus, we discuss the main neurotransmitters, receptors, and pathways that have been thought to play a role in l-dopa-induced dyskinesias from the perspective of basic neuroscience studies. Some important advances in deciphering the molecular pathways involved in these abnormal movements have occurred in recent years to reveal potential targets that could be used for therapeutic purposes. However, it has not been an easy road because there have been a plethora of components involved in the generation of these undesired movements, even bypassing the traditional and well-accepted dopamine receptor activation, as recently revealed by optogenetics. Here, we attempt to unify the available data with the hope of guiding and fostering future research in the field of striatal activation and abnormal movement generation. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Ledia F Hernández
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Jose A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Paolo Calabresi
- Fondazione Santa Lucia, IRCCS, Rome, Italy.,Clinica Neurologica, Università degli studi di Perugia, Ospedale Santa Maria della Misericordia, Perugia, Italy
| |
Collapse
|
11
|
Guerram M, Zhang LY, Jiang ZZ. G-protein coupled receptors as therapeutic targets for neurodegenerative and cerebrovascular diseases. Neurochem Int 2016; 101:1-14. [PMID: 27620813 DOI: 10.1016/j.neuint.2016.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 12/24/2022]
Abstract
Neurodegenerative and cerebrovascular diseases are frequent in elderly populations and comprise primarily of dementia (mainly Alzheimer's disease) Parkinson's disease and stroke. These neurological disorders (NDs) occur as a result of neurodegenerative processes and represent one of the most frequent causes of death and disability worldwide with a significant clinical and socio-economic impact. Although NDs have been characterized for many years, the exact molecular mechanisms that govern these pathologies or why they target specific individuals and specific neuronal populations remain unclear. As research progresses, many similarities appear which relate these diseases to one another on a subcellular level. Discovering these similarities offers hope for therapeutic advances that could ameliorate the conditions of many diseases simultaneously. G-protein coupled receptors (GPCRs) are the most abundant receptor type in the central nervous system and are linked to complex downstream pathways, manipulation of which may have therapeutic application in many NDs. This review will highlight the potential use of neurotransmitter GPCRs as emerging therapeutic targets for neurodegenerative and cerebrovascular diseases.
Collapse
Affiliation(s)
- Mounia Guerram
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Faculty of Exact Sciences and Nature and Life Sciences, Department of Biology, Larbi Ben M'hidi University, Oum El Bouaghi 04000, Algeria
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Hassan A, Heckman MG, Ahlskog JE, Wszolek ZK, Serie DJ, Uitti RJ, van Gerpen JA, Okun MS, Rayaprolu S, Ross OA. Association of Parkinson disease age of onset with DRD2, DRD3 and GRIN2B polymorphisms. Parkinsonism Relat Disord 2015; 22:102-5. [PMID: 26627941 DOI: 10.1016/j.parkreldis.2015.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/26/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Dopamine and glutamate are crucial neurotransmitters in Parkinson disease (PD). While recent large meta-analyses reported that genetic variation of dopamine (DRD2, DRD3) and glutamine (NMDA, GRIN2B) neurotransmitter receptors was not associated with PD risk, they could conceivably influence PD phenotype. We studied the association of these receptor polymorphisms relating to PD age of onset. METHODS There were 664 PD patients and 718 controls, all Caucasian, with stored DNA at Mayo Clinic, Jacksonville, Florida. Genotyping was performed for DRD2 (Taq 1A, rs1800497), DRD3 (rs6280), and NMDA (GRIN2B, rs7301328) polymorphisms with ABI Taqman assays. Single nucleotide polymorphism associations with age of onset were evaluated using dominant, recessive, and additive genotypic models. RESULTS DRD3 variant carriers had an approximate 4.4-year decrease in mean age of onset when both copies of the minor allele were present (P = 0.0034) and an approximate 1.5-year decrease in mean age at onset for every additional minor allele (P = 0.023) (recessive and additive models, respectively). There was no association with age of onset for DRD2 or GRIN2B under any statistical model (all P ≥ 0.22). CONCLUSIONS The DRD3 (rs6280) polymorphism, but not DRD2 (Taq1A) or GRIN2B, influences younger PD age of onset in the US Caucasian population. Validation of these findings in larger studies with other ethnic groups is indicated.
Collapse
Affiliation(s)
- Anhar Hassan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| | - Michael G Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | - J E Ahlskog
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Daniel J Serie
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Michael S Okun
- Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL, USA
| | - Sruti Rayaprolu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
13
|
Behaviour and prefrontal protein differences in C57BL/6N and 129 X1/SvJ mice. Brain Res Bull 2015; 116:16-24. [PMID: 26003851 DOI: 10.1016/j.brainresbull.2015.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/24/2023]
Abstract
Experimental animals provide valuable opportunities to establish aetiological mechanisms and test new treatments for neurodevelopmental psychiatric conditions. However, it is increasingly appreciated that inter-strain differences cannot be neglected in the experimental design. In addition, the importance of including females in preclinical - but also clinical - research is now recognised. Here, we compared behaviour and prefrontal protein differences in male and female C57BL/6N and 129X1/SvJ mice as both are commonly used experimental rodents. Relative to 129X1/SvJ mice, both sexes of C57BL/6N mice had weaker sensorimotor gating, measured in the prepulse inhibition (PPI) of startle paradigm, and were more sensitive to amphetamine challenge in the open field. The pattern of protein expression in the prefrontal cortex of C57BL6N mice was also clearly distinct from 129X1/SvJ mice. Proteins differentially expressed were those associated with oxidative metabolism, receptor protein signalling, cell communication and signal transduction and energy pathways. We suggest that the C57BL/6N mouse may usefully proxy features of the neurodevelopmental disorders and could have application in pre-translational screening of new therapeutic approaches. The 129X1/SvJ strain in contrast, might be better suited to experimental studies of causal risk factors expected to lower PPI and increase amphetamine sensitivity.
Collapse
|
14
|
Simms SL, Huettner DP, Kortagere S. In vivo characterization of a novel dopamine D3 receptor agonist to treat motor symptoms of Parkinson's disease. Neuropharmacology 2015; 100:106-15. [PMID: 25896768 DOI: 10.1016/j.neuropharm.2015.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/17/2015] [Accepted: 04/07/2015] [Indexed: 02/06/2023]
Abstract
Synthetic dopaminergic agents have found utility in treating neurological and neuropsychiatric disorders since the beginning of 19th century. The discovery of Levodopa (l-dopa) to effectively treat motor symptoms of Parkinson's disease (PD) revolutionized the therapy and remains a gold standard for treating PD. However, l-dopa therapy has been implicated in worsening of the non-motor symptoms including cognition and long-term therapy leads to plasticity and development of abnormal involuntary movements (AIMs) that are collectively called l-dopa induced dyskinesias (LID). Studies in rodents and non-human primates with PD have supported a role for dopamine D3 receptors in the etiology of both the motor symptoms and LID. We have recently developed SK609, a selective dopamine D3 receptor agonist with atypical signaling properties. In this study, we further characterized this novel small molecule using the unilateral lesioned rodent model of PD. In the forepaw stepping test paradigm, SK609 significantly improved the performance of the impaired paw and also normalized the bilateral asymmetry associated with the hemiparkinson rat. In addition, a chronic treatment of SK609 did not induce any AIMs and when used adjuvantly with l-dopa significantly reduced AIMs induced by l-dopa. Further, an optimal dose combination of SK609 with l-dopa was determined by dose dependent titrations of both SK609 and l-dopa that produced minimal AIMs and maximized the effect on improving motor symptoms. Results from this study suggest that SK609 is a novel dopaminergic agent that has the therapeutic potential to treat PD and LID. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Sherise L Simms
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Daniel P Huettner
- Department of Microbiology and Immunology, Centers for Molecular Parasitology, Virology and Translational Neuroscience, Institute for Molecular Medicine, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Centers for Molecular Parasitology, Virology and Translational Neuroscience, Institute for Molecular Medicine, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
15
|
Guridi J, Obeso JA. Letter to the Editor: Parkinson's disease and early subthalamotomy. J Neurosurg 2015; 122:980-1. [DOI: 10.3171/2014.8.jns141799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
16
|
Weber AM, Soreni N, Stanley JA, Greco A, Mendlowitz S, Szatmari P, Schachar R, Mannasis K, Pires P, Swinson R, Noseworthy MD. Proton magnetic resonance spectroscopy of prefrontal white matter in psychotropic naïve children and adolescents with obsessive-compulsive disorder. Psychiatry Res 2014; 222:67-74. [PMID: 24602517 DOI: 10.1016/j.pscychresns.2014.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 12/13/2013] [Accepted: 02/07/2014] [Indexed: 10/25/2022]
Abstract
Obsessive-compulsive disorder (OCD) has a typical onset during childhood or adolescence. Although recent in-vivo proton magnetic resonance spectroscopy ((1)H-MRS) studies report gray matter metabolite abnormalities in children and adolescents with OCD, there are no existing (1)H-MRS studies that measure white matter (WM) metabolite levels in this population. In the present study, we measured metabolite levels in the left and right prefrontal WM (LPFWM and RPFWM, respectively) of psychotropic-naïve children and adolescents with OCD (LPFWM: N=15, mean age 13.3±2.4 years; right RPFWM: N=14, mean age 13.0±2.3 years) and healthy controls (LPFWM: N=17, mean age 11.8±2.7 years; RPFWM: N=18, mean age 12.2±2.8 years). Spectra were acquired using a 3T single voxel PRESS sequence (1.5×2.0×2.0cm(3)). When age and sex effects were controlled, OCD patients had higher levels of RPFWM choline and N-acetyl-aspartate (NAA). In addition, RPFWM levels of NAA, creatine and myo-inositol were positively and significantly correlated with severity of OCD symptoms. In summary, this is the first published study of WM metabolite levels in children and adolescents with OCD. Our preliminary findings lend further support to the previous findings of WM abnormalities in OCD.
Collapse
Affiliation(s)
| | - Noam Soreni
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; Department of Psychiatry and Behavioural Neuroscience, Hamilton, ON, Canada; Offord Centre for Child Studies, McMaster University, McMaster Children׳s Hospital, Hamilton, ON, Canada; Anxiety Treatment and Research Center, St. Joseph׳s Healthcare, Hamilton, ON, Canada.
| | - Jeffrey A Stanley
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, School of Medicine, Detroit, MI, USA
| | - Alessia Greco
- Department of Psychiatry and Behavioural Neuroscience, Hamilton, ON, Canada; Offord Centre for Child Studies, McMaster University, McMaster Children׳s Hospital, Hamilton, ON, Canada
| | - Sandra Mendlowitz
- Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada
| | - Peter Szatmari
- Department of Psychiatry and Behavioural Neuroscience, Hamilton, ON, Canada; Offord Centre for Child Studies, McMaster University, McMaster Children׳s Hospital, Hamilton, ON, Canada
| | - Russell Schachar
- Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada
| | - Katharina Mannasis
- Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada
| | - Paulo Pires
- Department of Psychiatry and Behavioural Neuroscience, Hamilton, ON, Canada; Offord Centre for Child Studies, McMaster University, McMaster Children׳s Hospital, Hamilton, ON, Canada
| | - Richard Swinson
- Department of Psychiatry and Behavioural Neuroscience, Hamilton, ON, Canada; Offord Centre for Child Studies, McMaster University, McMaster Children׳s Hospital, Hamilton, ON, Canada; Anxiety Treatment and Research Center, St. Joseph׳s Healthcare, Hamilton, ON, Canada
| | - Michael D Noseworthy
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; Electrical & Computer Engineering, McMaster University, Hamilton, ON, Canada; Medical Physics & Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada; Diagnostic Imaging, St. Joseph׳s Healthcare, Hamilton, ON, Canada; Department of Radiology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
17
|
Picconi B, Calabresi P. Targeting metabotropic glutamate receptors as a new strategy against levodopa-induced dyskinesia in Parkinson's disease? Mov Disord 2014; 29:715-9. [PMID: 24591264 DOI: 10.1002/mds.25851] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/18/2013] [Accepted: 02/06/2014] [Indexed: 11/10/2022] Open
Abstract
Levodopa-induced dyskinesias (LIDs) represent one major motor disability of Parkinson's disease (PD) therapy. Thus, research effort is still devoted to finding agents that may improve parkinsonism and concomitantly reduce or avoid dyskinesia. Rodent and nonhuman primate models provide useful tools to study the molecular and neuronal bases of LIDs. Among the various strategies investigated recently, the use of drugs targeting metabotropic glutamate receptors has received large attention. In particular, use of antagonists of the subtype 5 of metabotropic glutamate receptors revealed promising preclinical and clinical results.
Collapse
|
18
|
Perfeito R, Cunha-Oliveira T, Rego AC. Reprint of: revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease-resemblance to the effect of amphetamine drugs of abuse. Free Radic Biol Med 2013; 62:186-201. [PMID: 23743292 DOI: 10.1016/j.freeradbiomed.2013.05.042] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a chronic and progressive neurological disease associated with a loss of dopaminergic neurons. In most cases the disease is sporadic but genetically inherited cases also exist. One of the major pathological features of PD is the presence of aggregates that localize in neuronal cytoplasm as Lewy bodies, mainly composed of α-synuclein (α-syn) and ubiquitin. The selective degeneration of dopaminergic neurons suggests that dopamine itself may contribute to the neurodegenerative process in PD. Furthermore, mitochondrial dysfunction and oxidative stress constitute key pathogenic events of this disorder. Thus, in this review we give an actual perspective to classical pathways involving these two mechanisms of neurodegeneration, including the role of dopamine in sporadic and familial PD, as well as in the case of abuse of amphetamine-type drugs. Mutations in genes related to familial PD causing autosomal dominant or recessive forms may also have crucial effects on mitochondrial morphology, function, and oxidative stress. Environmental factors, such as MPTP and rotenone, have been reported to induce selective degeneration of the nigrostriatal pathways leading to α-syn-positive inclusions, possibly by inhibiting mitochondrial complex I of the respiratory chain and subsequently increasing oxidative stress. Recently, increased risk for PD was found in amphetamine users. Amphetamine drugs have effects similar to those of other environmental factors for PD, because long-term exposure to these drugs leads to dopamine depletion. Moreover, amphetamine neurotoxicity involves α-syn aggregation, mitochondrial dysfunction, and oxidative stress. Therefore, dopamine and related oxidative stress, as well as mitochondrial dysfunction, seem to be common links between PD and amphetamine neurotoxicity.
Collapse
Affiliation(s)
- Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| |
Collapse
|
19
|
Opposite roles of NMDA receptors in relapsing and primary progressive multiple sclerosis. PLoS One 2013; 8:e67357. [PMID: 23840674 PMCID: PMC3696106 DOI: 10.1371/journal.pone.0067357] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 05/16/2013] [Indexed: 01/08/2023] Open
Abstract
Synaptic transmission and plasticity mediated by NMDA receptors (NMDARs) could modulate the severity of multiple sclerosis (MS). Here the role of NMDARs in MS was first explored in 691 subjects carrying specific allelic variants of the NR1 subunit gene or of the NR2B subunit gene of this glutamate receptor. The analysis was replicated for significant SNPs in an independent sample of 1548 MS subjects. The C allele of rs4880213 was found to be associated with reduced NMDAR-mediated cortical excitability, and with increased probability of having more disability than the CT/TT MS subjects. MS severity was higher in the CC group among relapsing-remitting MS (RR-MS) patients, while primary progressive MS (PP-MS) subjects homozygous for the T allele had more pronounced clinical worsening. Mean time to first relapse, but not to an active MRI scan, was lower in the CC group of RR-MS patients, and the number of subjects with two or more clinical relapses in the first two years of the disease was higher in CC compared to CT/TT group. Furthermore, the percentage of relapses associated with residual disability was lower in subjects carrying the T allele. Lesion load at the MRI was conversely unaffected by the C or T allele of this SNP in RR-MS patients. Axonal and neuronal degeneration at the optical coherence tomography was more severe in the TT group of PP-MS patients, while reduced retinal nerve fiber thickness had less consequences on visual acuity in RR-MS patients bearing the T allele. Finally, the T allele was associated with preserved cognitive abilities at the Rao's brief repeatable neuropsychological battery in RR-MS. Signaling through glutamate NMDARs enhances both compensatory synaptic plasticity and excitotoxic neurodegeneration, impacting in opposite ways on RR-MS and PP-MS pathophysiological mechanisms.
Collapse
|
20
|
Bogenpohl J, Galvan A, Hu X, Wichmann T, Smith Y. Metabotropic glutamate receptor 4 in the basal ganglia of parkinsonian monkeys: ultrastructural localization and electrophysiological effects of activation in the striatopallidal complex. Neuropharmacology 2013; 66:242-52. [PMID: 22634360 PMCID: PMC3490034 DOI: 10.1016/j.neuropharm.2012.05.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 03/14/2012] [Accepted: 05/13/2012] [Indexed: 11/23/2022]
Abstract
Group III metabotropic glutamate receptors (mGluR4,7,8) are widely distributed in the basal ganglia. Injection of group III mGluR agonists into the striatopallidal complex alleviates parkinsonian symptoms in 6-hydroxydopamine-treated rats. In vitro rodent studies have suggested that this may be partly due to modulation of synaptic transmission at striatopallidal and corticostriatal synapses through mGluR4 activation. However, the in vivo electrophysiological effects of group III mGluRs activation upon basal ganglia neurons activity in nonhuman primates remain unknown. Thus, in order to examine the anatomical substrates and physiological effects of group III mGluRs activation upon striatal and pallidal neurons in monkeys, we used electron microscopy immunohistochemistry to localize mGluR4, combined with local administration of the group III mGluR agonist L-AP4, or the mGluR4 positive allosteric modulator VU0155041, to assess the effects of group III mGluR activation on the firing rate and pattern of striatal and pallidal neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated parkinsonian monkeys. At the ultrastructural level, striatal mGluR4 immunoreactivity was localized in pre- (60%) and post-synaptic (30%) elements, while in the GPe, mGluR4 was mainly expressed pre-synaptically (90%). In the putamen, terminals expressing mGluR4 were evenly split between putative excitatory and inhibitory terminals, while in the GPe, most labeled terminals displayed the ultrastructural features of striatal-like inhibitory terminals, though putative excitatory boutons were also labeled. No significant difference was found between normal and parkinsonian monkeys. Extracellular recordings in awake MPTP-treated monkeys revealed that local microinjections of small volumes of L-AP4 resulted in increased firing rates in one half of striatal cells and one third of pallidal cells, while a significant number of neurons in both structures showed either opposite effects, or did not display any significant rate changes following L-AP4 application. VU0155041 administration had little effect on firing rates. Both compounds also had subtle effects on bursting and oscillatory properties, acting to increase the irregularity of firing. The occurrence of pauses in firing was reduced in the majority (80%) of GPe neurons after L-AP4 injection. Our findings indicate that glutamate can mediate multifarious physiological effects upon striatal and pallidal neurons through activation of pre-synaptic group III mGluRs at inhibitory and excitatory synapses in parkinsonian monkeys. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- James Bogenpohl
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329 USA
- NIH UDALL Center of Excellence for Parkinson’s Disease Research, Emory University, Atlanta, GA 30322 USA
| | - Adriana Galvan
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322 USA
- NIH UDALL Center of Excellence for Parkinson’s Disease Research, Emory University, Atlanta, GA 30322 USA
| | - Xing Hu
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329 USA
- NIH UDALL Center of Excellence for Parkinson’s Disease Research, Emory University, Atlanta, GA 30322 USA
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322 USA
- NIH UDALL Center of Excellence for Parkinson’s Disease Research, Emory University, Atlanta, GA 30322 USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322 USA
- NIH UDALL Center of Excellence for Parkinson’s Disease Research, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|
21
|
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
|
22
|
Perfeito R, Cunha-Oliveira T, Rego AC. Revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease--resemblance to the effect of amphetamine drugs of abuse. Free Radic Biol Med 2012; 53:1791-806. [PMID: 22967820 DOI: 10.1016/j.freeradbiomed.2012.08.569] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a chronic and progressive neurological disease associated with a loss of dopaminergic neurons. In most cases the disease is sporadic but genetically inherited cases also exist. One of the major pathological features of PD is the presence of aggregates that localize in neuronal cytoplasm as Lewy bodies, mainly composed of α-synuclein (α-syn) and ubiquitin. The selective degeneration of dopaminergic neurons suggests that dopamine itself may contribute to the neurodegenerative process in PD. Furthermore, mitochondrial dysfunction and oxidative stress constitute key pathogenic events of this disorder. Thus, in this review we give an actual perspective to classical pathways involving these two mechanisms of neurodegeneration, including the role of dopamine in sporadic and familial PD, as well as in the case of abuse of amphetamine-type drugs. Mutations in genes related to familial PD causing autosomal dominant or recessive forms may also have crucial effects on mitochondrial morphology, function, and oxidative stress. Environmental factors, such as MPTP and rotenone, have been reported to induce selective degeneration of the nigrostriatal pathways leading to α-syn-positive inclusions, possibly by inhibiting mitochondrial complex I of the respiratory chain and subsequently increasing oxidative stress. Recently, increased risk for PD was found in amphetamine users. Amphetamine drugs have effects similar to those of other environmental factors for PD, because long-term exposure to these drugs leads to dopamine depletion. Moreover, amphetamine neurotoxicity involves α-syn aggregation, mitochondrial dysfunction, and oxidative stress. Therefore, dopamine and related oxidative stress, as well as mitochondrial dysfunction, seem to be common links between PD and amphetamine neurotoxicity.
Collapse
Affiliation(s)
- Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | | |
Collapse
|
23
|
Amalric M, Lopez S, Goudet C, Fisone G, Battaglia G, Nicoletti F, Pin JP, Acher FC. Group III and subtype 4 metabotropic glutamate receptor agonists: discovery and pathophysiological applications in Parkinson's disease. Neuropharmacology 2012; 66:53-64. [PMID: 22664304 DOI: 10.1016/j.neuropharm.2012.05.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/28/2012] [Accepted: 05/21/2012] [Indexed: 12/22/2022]
Abstract
Restoring the balance between excitatory and inhibitory circuits in the basal ganglia, following the loss of dopaminergic (DA) neurons of the substantia nigra pars compacta, represents a major challenge to treat patients affected by Parkinson's disease (PD). The imbalanced situation in favor of excitation in the disease state may also accelerate excitotoxic processes, thereby representing a potential target for neuroprotective therapies. Reducing the excitatory action of glutamate, the major excitatory neurotransmitter in the basal ganglia, should lead to symptomatic improvement for PD patients and may promote the survival of DA neurons. Recent studies have focused on the modulatory action of metabotropic glutamate (mGlu) receptors on neurodegenerative diseases including PD. Group III mGlu receptors, including subtypes 4, 7 and 8, are largely expressed in the basal ganglia. Recent studies highlight the use of selective mGlu4 receptor positive allosteric modulators (PAMs) for the treatment of PD. Here we review the effects of newly-designed group-III orthosteric agonists on neuroprotection, neurorestoration and reduction of l-DOPA induced dyskinesia in animal models of PD. The combination of orthosteric mGlu4 receptor selective agonists with PAMs may open new avenues for the symptomatic treatment of PD. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- M Amalric
- Aix-Marseille University, CNRS UMR 7291, Laboratoire de Neurosciences Fonctionnelles, Case C, 3 Place Victor Hugo, 13331 Marseille Cedex 3, France.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Salvatore MF, Davis RW, Arnold JC, Chotibut T. Transient striatal GLT-1 blockade increases EAAC1 expression, glutamate reuptake, and decreases tyrosine hydroxylase phosphorylation at ser(19). Exp Neurol 2012; 234:428-36. [PMID: 22285253 DOI: 10.1016/j.expneurol.2012.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/24/2011] [Accepted: 01/10/2012] [Indexed: 12/16/2022]
Abstract
Three glutamate transporters, GLT-1, GLAST, and EAAC1, are expressed in striatum. GLT-1 and, to a lesser extent, GLAST are thought to play a primary role in glutamate reuptake and mitigate excitoxicity. Progressive tyrosine hydroxylase (TH) loss seen in Parkinson's disease (PD) is associated with increased extracellular glutamate. Glutamate receptor antagonists reduce nigrostriatal loss in PD models. These observations suggest that excess synaptic glutamate contributes to nigrostriatal neuron loss seen in PD. Decreased GLT-1 expression occurs in neurodegenerative disease and PD models, suggesting decreased GLT-1-mediated glutamate reuptake contributes to excitotoxicity. To determine how transient GLT-1 blockade affects glutamate reuptake dynamics and a Ca(2+)-dependent process in nigrostriatal terminals, ser(19) phosphorylation of TH, the GLT-1 inhibitor dihydrokainic acid (DHK) was delivered unilaterally to striatum in vivo and glutamate reuptake was quantified ex vivo in crude synaptosomes 3h later. Ca(2+)-influx is associated with excitotoxic conditions. The phosphorylation of TH at ser(19) is Ca(2+)-dependent, and a change resulting from GLT-1 blockade may signify the potential for excitotoxicity to nigrostriatal neurons. Synaptosomes from DHK infused striatum had a 43% increase in glutamate reuptake in conjunction with decreased ser(19) TH phosphorylation. Using a novel GLAST inhibitor and DHK, we determined that the GLAST-mediated component of increased glutamate reuptake increased 3-fold with no change in GLAST or GLT-1 protein expression. However, GLT-1 blockade increased EAAC1 protein expression ~20%. Taken together, these results suggest that GLT-1 blockade produces a transient increase in GLAST-mediated reuptake and EAAC1 expression coupled with reduced ser(19) TH phosphorylation. These responses could represent an endogenous defense against excitotoxicity to the nigrostriatal pathway.
Collapse
Affiliation(s)
- Michael F Salvatore
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130, USA.
| | | | | | | |
Collapse
|
25
|
Nicolás MJ, López-Azcárate J, Valencia M, Alegre M, Pérez-Alcázar M, Iriarte J, Artieda J. Ketamine-induced oscillations in the motor circuit of the rat basal ganglia. PLoS One 2011; 6:e21814. [PMID: 21829443 PMCID: PMC3146469 DOI: 10.1371/journal.pone.0021814] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 06/13/2011] [Indexed: 11/19/2022] Open
Abstract
Oscillatory activity can be widely recorded in the cortex and basal ganglia. This activity may play a role not only in the physiology of movement, perception and cognition, but also in the pathophysiology of psychiatric and neurological diseases like schizophrenia or Parkinson's disease. Ketamine administration has been shown to cause an increase in gamma activity in cortical and subcortical structures, and an increase in 150 Hz oscillations in the nucleus accumbens in healthy rats, together with hyperlocomotion.We recorded local field potentials from motor cortex, caudate-putamen (CPU), substantia nigra pars reticulata (SNr) and subthalamic nucleus (STN) in 20 awake rats before and after the administration of ketamine at three different subanesthetic doses (10, 25 and 50 mg/Kg), and saline as control condition. Motor behavior was semiautomatically quantified by custom-made software specifically developed for this setting.Ketamine induced coherent oscillations in low gamma (~ 50 Hz), high gamma (~ 80 Hz) and high frequency (HFO, ~ 150 Hz) bands, with different behavior in the four structures studied. While oscillatory activity at these three peaks was widespread across all structures, interactions showed a different pattern for each frequency band. Imaginary coherence at 150 Hz was maximum between motor cortex and the different basal ganglia nuclei, while low gamma coherence connected motor cortex with CPU and high gamma coherence was more constrained to the basal ganglia nuclei. Power at three bands correlated with the motor activity of the animal, but only coherence values in the HFO and high gamma range correlated with movement. Interactions in the low gamma band did not show a direct relationship to movement.These results suggest that the motor effects of ketamine administration may be primarily mediated by the induction of coherent widespread high-frequency activity in the motor circuit of the basal ganglia, together with a frequency-specific pattern of connectivity among the structures analyzed.
Collapse
Affiliation(s)
- María Jesús Nicolás
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
| | - Jon López-Azcárate
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
| | - Miguel Valencia
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
| | - Manuel Alegre
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
- Clinical Neurophysiology Section, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marta Pérez-Alcázar
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
| | - Jorge Iriarte
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
- Clinical Neurophysiology Section, Clínica Universidad de Navarra, Pamplona, Spain
| | - Julio Artieda
- Neurophysiology Laboratory, Neurosciences Area, CIMA, University of Navarra, Pamplona, Spain
- Clinical Neurophysiology Section, Clínica Universidad de Navarra, Pamplona, Spain
- * E-mail:
| |
Collapse
|
26
|
Mori F, Ribolsi M, Kusayanagi H, Siracusano A, Mantovani V, Marasco E, Bernardi G, Centonze D. Genetic variants of the NMDA receptor influence cortical excitability and plasticity in humans. J Neurophysiol 2011; 106:1637-43. [PMID: 21753020 DOI: 10.1152/jn.00318.2011] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
N-methyl-d-aspartate (NMDA) receptors play crucial roles in glutamate-mediated synaptic transmission and plasticity and are involved in a variety of brain functions. Specific single nucleotide polymorphisms (SNPs) in the genes encoding NMDA receptor subunits have been associated with some neuropsychiatric disorders involving altered glutamate transmission, but how these polymorphisms impact on synaptic function in humans is unknown. Here, the role of NMDA receptors in the control of cortical excitability and plasticity was explored by comparing the response to single, paired, and repetitive transcranial magnetic stimulations of the motor cortex in 77 healthy subjects carrying specific allelic variants of the NR1 subunit gene (GRIN1 rs4880213 and rs6293) or of the NR2B subunit gene (GRIN2B rs7301328, rs3764028, and rs1805247). Our results showed that individuals homozygous for the T allele in the rs4880213 GRIN1 SNP had reduced intracortical inhibition, as expected for enhanced glutamatergic excitation in these subjects. Furthermore, individuals carrying the G allele in the rs1805247 GRIN2B SNP show greater intracortical facilitation and greater long-term potentiation-like cortical plasticity after intermittent -burst stimulation. Our results provide novel insights into the function of NMDA receptors in the human brain and might contribute to the clarification of the synaptic bases of severe neuropsychiatric disorders associated with defective glutamate transmission.
Collapse
Affiliation(s)
- Francesco Mori
- Clinica Neurologica, Dipartimento di Neuroscienze, Università Tor Vergata, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Price DL, Rockenstein E, Ubhi K, Phung V, MacLean-Lewis N, Askay D, Cartier A, Spencer B, Patrick C, Desplats P, Ellisman MH, Masliah E. Alterations in mGluR5 expression and signaling in Lewy body disease and in transgenic models of alpha-synucleinopathy--implications for excitotoxicity. PLoS One 2010; 5:e14020. [PMID: 21103359 PMCID: PMC2982819 DOI: 10.1371/journal.pone.0014020] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 10/19/2010] [Indexed: 12/21/2022] Open
Abstract
Dementia with Lewy bodies (DLB) and Parkinson's Disease (PD) are neurodegenerative disorders of the aging population characterized by the abnormal accumulation of alpha-synuclein (alpha-syn). Previous studies have suggested that excitotoxicity may contribute to neurodegeneration in these disorders, however the underlying mechanisms and their relationship to alpha-syn remain unclear. For this study we proposed that accumulation of alpha-syn might result in alterations in metabotropic glutamate receptors (mGluR), particularly mGluR5 which has been linked to deficits in murine models of PD. In this context, levels of mGluR5 were analyzed in the brains of PD and DLB human cases and alpha-syn transgenic (tg) mice and compared to age-matched, unimpaired controls, we report a 40% increase in the levels of mGluR5 and beta-arrestin immunoreactivity in the frontal cortex, hippocampus and putamen in DLB cases and in the putamen in PD cases. In the hippocampus, mGluR5 was more abundant in the CA3 region and co-localized with alpha-syn aggregates. Similarly, in the hippocampus and basal ganglia of alpha-syn tg mice, levels of mGluR5 were increased and mGluR5 and alpha-syn were co-localized and co-immunoprecipitated, suggesting that alpha-syn interferes with mGluR5 trafficking. The increased levels of mGluR5 were accompanied by a concomitant increase in the activation of downstream signaling components including ERK, Elk-1 and CREB. Consistent with the increased accumulation of alpha-syn and alterations in mGluR5 in cognitive- and motor-associated brain regions, these mice displayed impaired performance in the water maze and pole test, these behavioral alterations were reversed with the mGluR5 antagonist, MPEP. Taken together the results from study suggest that mGluR5 may directly interact with alpha-syn resulting in its over activation and that this over activation may contribute to excitotoxic cell death in select neuronal regions. These results highlight the therapeutic importance of mGluR5 antagonists in alpha-synucleinopathies.
Collapse
Affiliation(s)
- Diana L. Price
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
| | - Edward Rockenstein
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Kiren Ubhi
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Van Phung
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Natalie MacLean-Lewis
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - David Askay
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
| | - Anna Cartier
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Christina Patrick
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Paula Desplats
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Mark H. Ellisman
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
28
|
Effects of depressive-like behavior of rats on brain glutamate uptake. Neurochem Res 2010; 35:1164-71. [PMID: 20405205 DOI: 10.1007/s11064-010-0169-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2010] [Indexed: 12/30/2022]
Abstract
Learned helplessness paradigm is a widely accepted animal model of depressive-like behavior based on stress. Glutamatergic system is closely involved with the stress-neurotoxicity in the brain and recently it is pointed to have a relevant role in the pathophysiology of depression disorder. Glutamate uptake is the main mechanism to terminate the glutamatergic physiological activity and to neuroprotection against excitotoxicity. We investigated the profile of glutamate uptake in female rats submitted to the learned helplessness paradigm and to different classes of stress related to the paradigm, in slices of brain cortex, striatum and hippocampus. Glutamate uptake in slices of hippocampus differ between learned helplessness (LH) and non-learned helplessness (NLH) animals immediately persisting up to 21 days after the paradigm. In addition, there were a decrease of glutamate uptake in the three brain structures analyzed at 21 days after the paradigm for LH animals. These results may contribute to better understand the role of the glutamatergic system on the depressive-like behavior.
Collapse
|
29
|
T cell-microglial dialogue in Parkinson's disease and amyotrophic lateral sclerosis: are we listening? Trends Immunol 2009; 31:7-17. [PMID: 19879804 DOI: 10.1016/j.it.2009.09.003] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/04/2009] [Accepted: 09/08/2009] [Indexed: 01/08/2023]
Abstract
Neuroinflammation is a pathological hallmark in Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), and is characterized by activated microglia and infiltrating T cells at sites of neuronal injury. In PD and ALS, neurons do not die alone; neuronal injury is non-cell-autonomous and depends on a well-orchestrated dialogue in which neuronally secreted misfolded proteins activate microglia and initiate a self-propagating cycle of neurotoxicity. Diverse populations and phenotypes of CD4(+) T cells crosstalk with microglia, and depending on their activation status, influence this dialogue and promote neuroprotection or neurotoxicity. A greater understanding of the T cell population that mediates these effects, as well as the molecular signals involved should provide targets for neuroprotective immunomodulation to treat these devastating neurodegenerative diseases.
Collapse
|
30
|
Lorenc-Koci E, Gołembiowska K, Pietraszek M, Wardas J. Treatment with 1,2,3,4,-tetrahydroisoquinoline affects glutamate release in the striatum but not the binding of [3H]MK-801 to NMDA receptors in the dopaminergic structures of the rat brain. Pharmacol Rep 2009; 61:798-806. [DOI: 10.1016/s1734-1140(09)70135-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 09/24/2009] [Indexed: 10/25/2022]
|
31
|
Cuomo D, Martella G, Barabino E, Platania P, Vita D, Madeo G, Selvam C, Goudet C, Oueslati N, Pin JP, Acher F, Pisani A, Beurrier C, Melon C, Kerkerian-Le Goff L, Gubellini P. Metabotropic glutamate receptor subtype 4 selectively modulates both glutamate and GABA transmission in the striatum: implications for Parkinson’s disease treatment. J Neurochem 2009; 109:1096-105. [DOI: 10.1111/j.1471-4159.2009.06036.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
32
|
Takada-Takatori Y, Kume T, Izumi Y, Ohgi Y, Niidome T, Fujii T, Sugimoto H, Akaike A. Roles of Nicotinic Receptors in Acetylcholinesterase Inhibitor-Induced Neuroprotection and Nicotinic Receptor Up-Regulation. Biol Pharm Bull 2009; 32:318-24. [DOI: 10.1248/bpb.32.318] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuki Takada-Takatori
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Doshisha Women's College
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yasuhiko Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yuta Ohgi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Tetsuhiro Niidome
- Department of Neuroscience for Drug Discovery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Takeshi Fujii
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Doshisha Women's College
| | - Hachiro Sugimoto
- Department of Neuroscience for Drug Discovery Research, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Akinori Akaike
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
33
|
Chung E, Chen L, Chan Y, Yung K. Downregulation of glial glutamate transporters after dopamine denervation in the striatum of 6-hydroxydopamine-lesioned rats. J Comp Neurol 2008; 511:421-37. [DOI: 10.1002/cne.21852] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Martella G, Platania P, Vita D, Sciamanna G, Cuomo D, Tassone A, Tscherter A, Kitada T, Bonsi P, Shen J, Pisani A. Enhanced sensitivity to group II mGlu receptor activation at corticostriatal synapses in mice lacking the familial parkinsonism-linked genes PINK1 or Parkin. Exp Neurol 2008; 215:388-96. [PMID: 19071114 DOI: 10.1016/j.expneurol.2008.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 10/17/2008] [Accepted: 11/08/2008] [Indexed: 10/21/2022]
Abstract
An altered glutamatergic input at corticostriatal synapses has been shown in experimental models of Parkinson's disease (PD). In the present work, we analyzed the membrane and synaptic responses of striatal neurons to metabotropic glutamate (mGlu) receptor activation in two different mouse models of inherited PD, linked to mutations in PINK1 or Parkin genes. Both in PINK1 and Parkin knockout ((-/-)) mice, activation of group I mGlu receptors by 3,5-DHPG caused a membrane depolarization coupled to an increase in firing frequency in striatal cholinergic interneurons that was comparable to the response observed in the respective wild-type (WT) interneurons. The sensitivity to group II and III mGlu receptors was tested on cortically-evoked excitatory postsynaptic potentials (EPSPs) recorded from medium spiny neurons (MSNs). Both LY379268 and L-AP4, agonists for group II and III, respectively, had no effect on intrinsic membrane properties, but dose-dependently reduced the amplitude of corticostriatal EPSPs. However, both in PINK1(-/-) and Parkin(-/-) mice, LY379268, but not L-AP4, exhibited a greater potency as compared to WT in depressing EPSP amplitude. Accordingly, the dose-response curve for the response to LY379268 in both knockout mice was shifted leftward. Moreover, consistent with a presynaptic site of action, both LY379268 and L-AP4 increased the paired-pulse ratio either in PINK1(-/-) and Parkin(-/-) or in WT mice. Acute pretreatment with L-dopa did not rescue the enhanced sensitivity to LY379268. Together, these results suggest that the selective increase in sensitivity of striatal group II mGlu receptors represents an adaptive change in mice in which an altered dopamine metabolism has been documented.
Collapse
Affiliation(s)
- G Martella
- Department of Neuroscience, University Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Papa SM. The cannabinoid system in Parkinson's disease: multiple targets to motor effects. Exp Neurol 2008; 211:334-8. [PMID: 18433745 DOI: 10.1016/j.expneurol.2008.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 02/25/2008] [Accepted: 03/07/2008] [Indexed: 01/09/2023]
Affiliation(s)
- Stella M Papa
- Department of Neurology, Emory University School of Medicine, 6000 WMRC, 101 Woodruff Circle, Atlanta, Georgia, 30322, USA.
| |
Collapse
|
36
|
Rousseaux CG. A Review of Glutamate Receptors II: Pathophysiology and Pathology. J Toxicol Pathol 2008. [DOI: 10.1293/tox.21.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Colin G. Rousseaux
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa
| |
Collapse
|
37
|
Quintero JE, Day BK, Zhang Z, Grondin R, Stephens ML, Huettl P, Pomerleau F, Gash DM, Gerhardt GA. Amperometric measures of age-related changes in glutamate regulation in the cortex of rhesus monkeys. Exp Neurol 2007; 208:238-46. [PMID: 17927982 DOI: 10.1016/j.expneurol.2007.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 08/01/2007] [Accepted: 08/07/2007] [Indexed: 11/17/2022]
Abstract
l-glutamate (glutamate) is the principal excitatory neurotransmitter of the central nervous system and is involved in altered neural function during aging and in neurodegenerative diseases. Relatively little is known about the mechanisms of glutamate signaling in the primate brain, in part, because there is an absence of a method capable of rapidly measuring glutamate in either a non-clinical or a clinical setting. We have addressed this paucity of information by measuring extracellular glutamate at 1 Hz in the pre-motor and motor cortices of young, middle-aged, and aged monkeys using a minimally invasive amperometric recording method. In the motor cortex, mean resting glutamate levels were five times higher in the aged group compared to the young group while the pre-motor cortex showed an increasing trend in resting glutamate levels that was not statistically significant. In addition, we measured rapid, phasic glutamate release after local pressure-ejection of nanoliter volumes of either isotonic 70 mM potassium (to stimulate glutamate release) or 1 mM glutamate (to study glutamate uptake) into the pre-motor and motor cortex. In the pre-motor cortex, we measured reproducible glutamate uptake signals that had a significantly decreased (47%) rate of glutamate uptake in aged animals compared to young animals. However, following a 70 mM potassium delivery, we did not observe any consistent changes in evoked release between young versus aged animals. Using these non-clinical microelectrodes to measure glutamate signaling in the brain, our results support the hypothesis that the glutamatergic system undergoes reorganization with aging of the central nervous system.
Collapse
Affiliation(s)
- Jorge E Quintero
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0098, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yadava N, Nicholls DG. Spare respiratory capacity rather than oxidative stress regulates glutamate excitotoxicity after partial respiratory inhibition of mitochondrial complex I with rotenone. J Neurosci 2007; 27:7310-7. [PMID: 17611283 PMCID: PMC6794596 DOI: 10.1523/jneurosci.0212-07.2007] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Partial inhibition of mitochondrial respiratory complex I by rotenone reproduces aspects of Parkinson's disease in rodents. The hypothesis that rotenone enhancement of neuronal cell death is attributable to oxidative stress was tested in an acute glutamate excitotoxicity model using primary cultures of rat cerebellar granule neurons. As little as 5 nM rotenone increased mitochondrial superoxide (O2*-) levels and potentiated glutamate-induced cytoplasmic Ca2+ deregulation, the first irreversible stage of necrotic cell death. However, the potent cell-permeant O2*- trap manganese tetrakis (N-ethylpyridinium-2yl) porphyrin failed to prevent the effects of the inhibitor. The bioenergetic consequences of rotenone addition were quantified by monitoring cell respiration. Glutamate activation of NMDA receptors used the full respiratory capacity of the in situ mitochondria, and >80% of the glutamate-stimulated respiration was attributable to increased cellular ATP demand. Rotenone at 20 nM inhibited basal and carbonyl cyanide p-trifluoromethoxyphenylhydrazone-stimulated cell respiration and caused respiratory failure in the presence of glutamate. ATP synthase inhibition by oligomycin was also toxic in the presence of glutamate. We conclude that the cell vulnerability in the rotenone model of partial complex I deficiency under these specific conditions is primarily determined by spare respiratory capacity rather than oxidative stress.
Collapse
|
39
|
Chung EKY, Chen LW, Chan YS, Yung KKL. Up-regulation in expression of vesicular glutamate transporter 3 in substantia nigra but not in striatum of 6-hydroxydopamine-lesioned rats. Neurosignals 2007; 15:238-48. [PMID: 17435391 DOI: 10.1159/000101704] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 02/12/2007] [Indexed: 11/19/2022] Open
Abstract
Overactivity of the glutamatergic system is suggested to be closely related to the onset and pathogenesis of Parkinson's disease. Vesicular glutamate transporters (VGLUT1, T2 and T3) are a group of glutamate transporters in neurons that are responsible for transporting glutamate into synaptic vesicles and they are key elements for homeostasis of glutamate neurotransmission. The present study was aimed to investigate the expression of VGLUT1, T2 and T3 proteins after the onset of Parkinson's disease. A rat model of Parkinson's disease, the 6-hydroxydopamine-lesioned rat, was employed. Immunocytochemistry revealed that VGLUT1, T2 and T3 immunoreactivity was not modulated in the striatum of the lesioned rat. Western blotting analyses also showed that there was no change in the expression of T1, T2 and T3 proteins in the striatum. In contrast, no VGLUT1 protein was detected in the substantia nigra. After the lesion, levels of VGLUT2 immunoreactivity and protein were not modulated. Significant increase of VGLUT3 immunoreactivity was observed in the perikarya of GABAergic substantia nigra pars reticulata neurons (+14.7%) although VGLUT3 protein was not modulated in the nigral tissues. VGLUT3 in GABAergic neurons is suggested to play a role in GABA synthesis. The present results may therefore implicate that VGLUT1 and T2 are not modulated in the striatum and the substantia nigra of the 6-hydroxydopamine-lesioned rat and only VGLUT3 plays a role in pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- E K Y Chung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong, PR China
| | | | | | | |
Collapse
|
40
|
Avshalumov MV, Bao L, Patel JC, Rice ME. H2O2 signaling in the nigrostriatal dopamine pathway via ATP-sensitive potassium channels: issues and answers. Antioxid Redox Signal 2007; 9:219-31. [PMID: 17115944 DOI: 10.1089/ars.2007.9.219] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The role of reactive oxygen species (ROS) as signaling agents is increasingly appreciated. Studies of ROS functions in the central nervous system, however, are only in their infancy. Using fast-scan cyclic voltammetry and fluorescence imaging in brain slices, the authors discovered that hydrogen peroxide (H2O2) is an endogenous regulator of dopamine release in the dorsal striatum. Given the key role of dopamine in motor, reward, and cognitive pathways, regulation by H2O2 has implications for normal dopamine function, as well as for dysfunction of dopamine transmission. In this review, data are summarized to show that H2O2 is a diffusible messenger in the striatum, generated downstream from glutamate receptor activation, and an intracellular signal in dopamine neurons of the substantia nigra, generated during normal pacemaker activity. The mechanism by which H2O2 inhibits dopamine release and dopamine cell activity is activation of ATP-sensitive K+ (KATP) channels. Characteristics of the neuronal and glial antioxidant networks required to permit H2O2 signaling, yet prevent oxidative damage, are also considered. Lastly, estimates of physiological H2O2 levels are discussed, and strengths and limitations of currently available methods for H2O2 detection, including fluorescence imaging using dichlorofluorescein (DCF) and the next generation of fluorescent probes, are considered.
Collapse
Affiliation(s)
- Marat V Avshalumov
- Department of Neurosurgery and Department of Physiology and Neuroscience, New York University School of Medicine, New York 10016, USA
| | | | | | | |
Collapse
|
41
|
|
42
|
Abstract
The protection of neurons from damage and death in neurodegenerative disorders, such as Alzheimer disease (AD), is a major challenge for neuroscientists in the 21st century. The amyloid beta-protein plays an important role in the degenerative process of the disease and increases the vulnerability of cultured cortical neurons to glutamate neurotoxicity. Glutamate may, therefore, play an important role in amyloid beta-protein-induced cytotoxicity in the cerebral cortex. Results show that cholinesterase inhibitors such as donepezil protect cortical neurons against glutamate neurotoxicity via alpha4beta2 and alpha7 nicotinic acetylcholine receptors at least partly by inhibiting the process of apoptosis. Donepezil also protects against ischemic insults such as those seen in vascular dementia; however, this does not seem to be mediated by nicotinic receptors. This review summarizes data that suggest donepezil possesses neuroprotective actions in addition to amelioration of cognitive deficits by inhibition of acetylcholinesterase.
Collapse
Affiliation(s)
- Akinori Akaike
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
43
|
Gubellini P, Eusebio A, Oueslati A, Melon C, Kerkerian-Le Goff L, Salin P. Chronic high-frequency stimulation of the subthalamic nucleus and L-DOPA treatment in experimental parkinsonism: effects on motor behaviour and striatal glutamate transmission. Eur J Neurosci 2006; 24:1802-14. [PMID: 17004943 DOI: 10.1111/j.1460-9568.2006.05047.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Hyperactivity of striatal glutamatergic synaptic transmission in response to dopamine depletion plays a major role in the pathogenesis of parkinsonian motor symptoms. In the present study we investigated the impact, on this hyperactivity, of chronic dyskinesiogenic L-DOPA treatment, combined or not with high-frequency stimulation (HFS) of the subthalamic nucleus (STN). In vitro patch-clamp recordings were performed from striatal spiny neurons of hemiparkinsonian rats (intranigral 6-OHDA injection). Here we show that dyskinesiogenic L-DOPA treatment exacerbated striatal glutamatergic hyperactivity induced by 6-OHDA lesion. Chronic 5-day STN HFS had the opposite effect, reducing striatal glutamatergic transmission in both parkinsonian and dyskinetic animals. Consistently, chronic HFS stimulation could progressively ameliorate motor parkinsonian signs (akinesia) but, conversely, did not improve L-DOPA-induced dyskinesia (LID). Thus, the effects of L-DOPA and HFS on corticostriatal transmission seem to be dissociated. These data show for the first time that dyskinesiogenic L-DOPA treatment and chronic STN HFS with antiakinetic effects induce opposite plastic rearrangements in the striatum. The interaction between these two treatments provides further evidence that striatal glutamatergic hyperactivity is a pathophysiological correlate of akinesia rather than LID.
Collapse
Affiliation(s)
- Paolo Gubellini
- Equipe IC2N, Institut de Biologie du Développement de Marseille-Luminy (IBDML), UMR6216, 31, Chemin Joseph Aiguier CNRS-Université de la Méditerranée, 13402 Marseille, CEDEX 20, France.
| | | | | | | | | | | |
Collapse
|
44
|
Lui PW, Yeung CW, Yung WH, Shi Y, Chen LW, Chan YS, Yung KKL. Ablation of gene expression of N-methyl-D-aspartate receptor one by antisense oligonucleotides in striatal neurons in culture. Neurosignals 2006; 14:303-16. [PMID: 16772733 DOI: 10.1159/000093045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Accepted: 02/28/2006] [Indexed: 11/19/2022] Open
Abstract
In the present study, a twenty-mer antisense oligonucleotide specific for N-methyl-D-aspartate receptor one (ANR1) was applied to striatal neurons in primary cell culture. The ANR1 was found to be specific and nontoxic. Significant reductions in expression of NR1 mRNA and proteins were resulted after a single dose of ANR1 transcripts. Interestingly, there were reductions in total NR1 proteins but two phosphorylated forms of NR1 proteins at serine 896 and 897 residues were not reduced. There was also no change in the pattern of distribution of NR1 immunoreactivity in the striatal neurons. In addition, significant reductions of NMDA-mediated peak inward current were found after application of a higher concentration of ANR1 (20-100 microM) by patch clamp recordings. The present results indicate that ANR1 is a useful agent in reducing NMDA receptor functions. The present data thus provide detailed cellular and molecular mechanisms to explain our previous findings of amelioration of motor symptoms in a rat model of Parkinson's disease. More importantly, application of ANR1 was also found to display neuroprotective effects of striatal neurons against NMDA-induced excitotoxic cell death. The findings have implications in development of new approach in prevention of cell death in neurodegenerative diseases and new treatments for these diseases.
Collapse
Affiliation(s)
- P W Lui
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, PR China
| | | | | | | | | | | | | |
Collapse
|
45
|
Nyholm D. Pharmacokinetic optimisation in the treatment of Parkinson's disease : an update. Clin Pharmacokinet 2006; 45:109-36. [PMID: 16485914 DOI: 10.2165/00003088-200645020-00001] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Pharmacotherapy for Parkinson's disease is focused on dopaminergic drugs, mainly the dopamine precursor levodopa and dopamine receptor agonists. The elimination half-life (t(1/2)) of levodopa from plasma (in combination with a decarboxylase inhibitor) of about 1.5 hours becomes more influential as the disease progresses. The long-duration of response to levodopa, which is evident in early Parkinson's disease, diminishes and after a few years of treatment motor performance is closely correlated to the fluctuating plasma concentrations of levodopa. Absorption of levodopa in the proximal small intestine depends on gastric emptying, which is erratic and may be slowed in Parkinson's disease. The effects of levodopa on motor function are dependent on gastric emptying in patients in the advanced stages of disease. The current treatment concept is continuous dopaminergic stimulation (CDS). Sustained-release formulations of levodopa may provide more stable plasma concentrations. Oral liquid formulations shorten the time to reach peak concentration and onset of effect but do not affect plasma levodopa variability. The t(1/2) of levodopa can be prolonged by adding a catechol-O-methyltransferase inhibitor (entacapone or tolcapone), which may reduce fluctuations in plasma concentrations, although both peak and trough concentrations are increased with frequent administration. Intravenous and enteral (duodenal/jejunal) infusions of levodopa yield stable plasma levodopa concentrations and motor performance. Enteral infusion is feasible on a long-term basis in patients with severe fluctuations. Among the dopamine receptor agonists the ergot derivatives bromocriptine, cabergoline, dihydroergocryptine and pergolide, and the non-ergot derivatives piribedil, pramipexole and ropinirole, have longer t(1/2) compared with levodopa. Thus, they stimulate dopamine receptors in a less pulsatile manner, yet pharmacokinetic studies of repeated doses of dopamine receptor agonists are few. Optimisation of these drugs is often performed with standardised titration schedules. Apomorphine and lisuride have short t(1/2) and are suitable for subcutaneous infusion, with results similar to those of levodopa infusion. Transdermal administration of dopamine receptor agonists such as rotigotine might be an alternative in the future. In general, initial dopamine receptor agonist monotherapy is associated with poorer motor performance and lower incidence of motor complications compared with levodopa. Buccal administration of the monoamine oxidase-B inhibitor selegiline (deprenyl) provides better absorption and less formation of metabolites compared with standard tablets. To conclude, several new drugs, formulations and routes of administration have been introduced in the treatment of Parkinson's disease during the last decade, mainly with CDS as the aim. CDS can be approached by optimising the use of dopaminergic drugs based on pharmacokinetic data.
Collapse
Affiliation(s)
- Dag Nyholm
- Department of Neuroscience, Neurology, Uppsala University Hospital, Uppsala, Sweden.
| |
Collapse
|
46
|
Hasbani DM, Perez FA, Palmiter RD, O'Malley KL. Dopamine depletion does not protect against acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity in vivo. J Neurosci 2006; 25:9428-33. [PMID: 16221852 PMCID: PMC6725707 DOI: 10.1523/jneurosci.0130-05.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dopamine (DA) has been postulated to play a role in the loss of dopaminergic substantia nigra (SN) neurons in Parkinson's disease because of its propensity to oxidize and form quinones and other reactive oxygen species that can alter cellular function. Moreover, DA depletion can attenuate dopaminergic cell loss in vitro. To test the contribution of DA to SN impairment in vivo, we used DA-deficient mice, which lack the enzyme tyrosine hydroxylase in dopaminergic cells, and mice pharmacologically depleted of DA by alpha-methyl-p-tyrosine pretreatment. Mice were treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a toxin that produces parkinsonian pathology in humans, nonhuman primates, and rodents. In contrast to in vitro results, genetic or pharmacologic DA depletion did not attenuate loss of dopaminergic neurons in the SN or dopaminergic neuron terminals in the striatum. These results suggest that DA does not contribute to acute MPTP toxicity in vivo.
Collapse
Affiliation(s)
- Daphne M Hasbani
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
47
|
Blandini F. Neuroprotection by rasagiline: a new therapeutic approach to Parkinson's disease? CNS DRUG REVIEWS 2005; 11:183-94. [PMID: 16007239 PMCID: PMC6741719 DOI: 10.1111/j.1527-3458.2005.tb00269.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuronal death in Parkinson's disease (PD) may originate from the reciprocal interactions of a restricted number of conditions, such as mitochondrial defects, oxidative stress and protein mishandling, which would favor a state of apoptotic cell death in the nigrostriatal pathway. The search for pharmacological treatments able to counteract the nigrostriatal degeneration, possibly by interfering with these phenomena, has recently raised considerable interest in rasagiline [R(+)-N-propargyl-1-aminoindan], a potent, selective, and irreversible inhibitor of monoamine oxidase B (MAO-B). Rasagiline, like selegiline, is a propargylamine, but is approximately 10 times more potent. Unlike selegiline, rasagiline is not metabolized to amphetamine and/or methamphetamine and is devoid of sympathomimetic activity. Numerous experimental studies, conducted both in vitro and in vivo, have shown that rasagiline possesses significant protective properties on neuronal populations. The pro-survival effects of the drug appear to be linked to its propargyl moiety, rather than to the inhibitory effect on MAO-B. Rasagiline's major metabolite, aminoindan--which possesses intrinsic neuroprotective activity--may also contribute to the beneficial effects of the parent compound. Rasagiline has been recently evaluated in early PD patients, with results that are consistent with slowing the progression of the disease. Therefore, the neuroprotective activity shown by the drug under experimental conditions may be reflected in the clinic, thus providing new perspectives for the treatment of PD.
Collapse
Affiliation(s)
- Fabio Blandini
- Laboratory of Functional Neurochemistry, IRCCS Neurological Institute C. Mondino, Via Mondino, 2 27100 Pavia, Italy.
| |
Collapse
|
48
|
Samadi P, Grégoire L, Rassoulpour A, Guidetti P, Izzo E, Schwarcz R, Bédard PJ. Effect of kynurenine 3-hydroxylase inhibition on the dyskinetic and antiparkinsonian responses to levodopa in Parkinsonian monkeys. Mov Disord 2005; 20:792-802. [PMID: 15954116 DOI: 10.1002/mds.20596] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Homeostatic interactions between dopamine and glutamate are central to the normal physiology of the basal ganglia. This relationship is altered in Parkinsonism and in levodopa-induced dyskinesias (LID), resulting in an upregulation of corticostriatal glutamatergic function. Kynurenic acid (KYNA), a tryptophan metabolite with antagonist activity at ionotropic glutamate receptors and the capability to inhibit glutamate release presynaptically, might therefore be of therapeutic value in LID. To evaluate this hypothesis, we used a pharmacological tool, the kynurenine 3-hydroxylase inhibitor Ro 61-8048, which raises KYNA levels acutely. Ro 61-8048 was tested in MPTP cynomolgus monkeys with a stable parkinsonian syndrome and reproducible dyskinesias after each dose of levodopa. Serum and CSF concentrations of KYNA and its precursor kynurenine increased dose-dependently after Ro 61-8048 administration, alone or in combination with levodopa. Coadministration of Ro 61-8048 with levodopa produced a moderate but significant reduction in the severity of dyskinesias while maintaining the motor benefit. These results suggest that elevation of KYNA levels through inhibition of kynurenine 3-hydroxylase constitutes a promising novel approach for managing LID in Parkinson's disease.
Collapse
Affiliation(s)
- Pershia Samadi
- Centre de Recherche en Neurosciences, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Kress GJ, Reynolds IJ. Dopaminergic neurotoxins require excitotoxic stimulation in organotypic cultures. Neurobiol Dis 2005; 20:639-45. [PMID: 15996475 DOI: 10.1016/j.nbd.2005.04.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2004] [Revised: 04/09/2005] [Accepted: 04/26/2005] [Indexed: 10/25/2022] Open
Abstract
We have investigated the properties of the dopaminergic neurotoxins 6-hydroxydopamine, 1-methyl-4-phenylpyridinium and rotenone using an organotypic culture that included slices of substantia nigra, striatum and cortex maintained for about 20 days in vitro. At this age, the organotypic culture contains dopaminergic neurons, visualized using tyrosine hydroxylase (TH) immunohistochemistry, that project into the striatal slice and extend up to 1 mm into the cortical slice. Using TH immunohistochemistry to assess survival of dopaminergic neurons, we found that the three dopaminergic toxins alone were not selectively neurotoxic. However, the addition of a low concentration of N-methyl-d-aspartate together with each individual toxin resulted in profound injury to the dopaminergic neurons, reflected by the loss of cell bodies and the fragmentation of processes. The combined toxicity was completely blocked by MK801. To assess the specificity of the injury, we measured the diameter of cell nuclei in the organotypic culture stained with Hoechst 33342 because the nucleus shrinks when neurons are injured. These measurements showed that the combined toxin treatment selectively injured only the TH immunoreactive cells. Thus, in a model culture system where dopaminergic neurons innervate appropriate targets, excitotoxicity appears to be essential for the manifestation of the toxic actions of 6-hydroxydopamine, 1-methyl-4-phenylpyridinium and rotenone.
Collapse
Affiliation(s)
- Geraldine J Kress
- Department of Pharmacology, University of Pittsburgh, W1351 Biomedical Science Tower, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
50
|
Giustizieri M, Bernardi G, Mercuri NB, Berretta N. Distinct mechanisms of presynaptic inhibition at GABAergic synapses of the rat substantia nigra pars compacta. J Neurophysiol 2005; 94:1992-2003. [PMID: 15944237 DOI: 10.1152/jn.00171.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the mechanisms of presynaptic inhibition of GABAergic neurotransmission by group III metabotropic glutamate receptors (mGluRs) and GABA(B) receptors, in dopamine (DA) neurons of the substantia nigra pars compacta (SNc). Both the group III mGluRs agonist L-(+)-2-amino-4-phosphonobutyric acid (AP4, 100 microM) and the GABA(B) receptor agonist baclofen (10 microM) reversibly depressed the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) to 48.5 +/- 2.7 and 79.3 +/- 1.6% (means +/- SE) of control, respectively. On the contrary, the frequency of action potential-independent miniature IPSCs (mIPSCs), recorded in tetrodotoxin (TTX, 1 microM) and cadmium (100 microM) were insensitive to AP4 but were reduced by baclofen to 49.7 +/- 8.6% of control. When the contribution of voltage-dependent calcium channels (VDCCs) to synaptic transmission was boosted with external barium (1 mM), AP4 became effective in reducing TTX-resistant mIPSCs to 65.4 +/- 3.9% of control, thus confirming a mechanism of presynaptic inhibition involving modulation of VDCCs. Differently from AP4, baclofen inhibited to 58.5 +/- 6.7% of control the frequency mIPSCs recorded in TTX and the calcium ionophore ionomycin (2 microM), which promotes Ca2+-dependent, but VDCC-independent, transmitter release. Moreover, in the presence of alpha-latrotoxin (0.3 nM), to promote a Ca2+-independent vesicular release of GABA, baclofen reduced mIPSC frequency to 48.1 +/- 3.2% of control, while AP4 was ineffective. These results indicate that group III mGluRs depress GABA release to DA neurons of the SNc through inhibition of presynaptic VDCCs, while presynaptic GABA(B) receptors directly impair transmitter exocytosis.
Collapse
Affiliation(s)
- Michela Giustizieri
- Centro Europeo di Ricerca sul Cervello Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | | | | | | |
Collapse
|