1
|
Yan R, Zheng X, Yin Y, Zhang J, Cui Y, Su D, Wan Z, Feng T. Treatment for Dyskinesia in Parkinson's Disease: A Network Meta-analysis of Randomized Controlled Trials. Mov Disord 2025. [PMID: 40099430 DOI: 10.1002/mds.30179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Dyskinesia is a motor complication of Parkinson's disease (PD) posing therapeutic challenges. The optimal therapy for dyskinesia in PD has not been identified due to the lack of comprehensive evaluation of treatments. OBJECTIVE The aim was to compare the efficacy and safety of interventions for alleviating levodopa-induced dyskinesia in PD. METHODS We conducted a Bayesian network meta-analysis (NMA) by systematically searching PubMed, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, and EudraCT databases up to April 1, 2024. The primary efficacy outcome was the change in scores on dyskinesia rating scales from baseline. RESULTS The study included 85 randomized controlled trials (RCT) involving 13,826 PD patients, comprising 39 interventions. Nine treatments were significantly more effective in reducing scores on dyskinesia rating scales than control (placebo, sham surgery, sham repetitive transcranial magnetic stimulation, or best medical treatment). Globus pallidus interna deep brain stimulation (GPi-DBS) had the highest probability to be the most effective (standardized mean difference, 95% credible interval: -1.27, -1.65 to -0.88; surface under the cumulative ranking curve [SUCRA]: 97.4%), followed by levodopa-carbidopa intestinal gel infusion (SUCRA = 89.7%), subthalamic nucleus (STN)-DBS (SUCRA = 89%), immediate-release (IR) amantadine (SUCRA = 86.5%), pallidotomy (SUCRA = 84.9%), ADS-5102 (SUCRA = 82.9%), clozapine (SUCRA = 77.2%), OS320 (SUCRA = 64.8%), and AFQ056 (SUCRA = 54.5%). GPi-DBS was superior to STN-DBS, and pallidotomy ranked higher than subthalamotomy. ADS-5102 and OS320 had higher adverse event (AE) rates compared to control, whereas AFQ056 and ADS-5102 were linked to more serious AEs. CONCLUSIONS This RCT-based NMA identifies and ranks nine efficacious interventions for dyskinesia in PD. GPi-DBS may be the most effective therapy for treating dyskinesia, with IR amantadine ranking highest among oral medications. Novel anti-dyskinetic medications are associated with less-favorable tolerance profiles. © 2025 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rui Yan
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaoqing Zheng
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yixuan Yin
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junjiao Zhang
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yusha Cui
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dongning Su
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhirong Wan
- Department of Neurology, Aerospace Center Hospital, Beijing, China
| | - Tao Feng
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
2
|
Zou ZZ, Han MJ, Chang Y, Li G. Insights into the Metabolism, Disposition, and Quantitative Profile of mGlu5 NAM AE90015 with Metabolite Identification and a Novel Integration Method. Molecules 2024; 29:5724. [PMID: 39683882 DOI: 10.3390/molecules29235724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
AE90015 is a highly specific and effective negative allosteric modulator (NAM) for the human mGlu5 receptor, showing significant promise for treating Parkinson's disease. An in vivo rat oral dose study was conducted on AE90015, which involved the collection of urine and bile samples over a 24 h period. At the study's endpoint, plasma, liver, brain, and renal tissues were also collected. A total of 30 metabolites of AE90015 were identified and structurally characterized or detected using high-resolution LC-MS/MSn. These metabolites fall into four categories: mono-hydroxyl, di-hydroxyl, mono-hydroxyl glucuronide, and di-hydroxyl glucuronide. This study provided a comprehensive overview of the metabolism, excretion, and disposition of AE90015, a promising NAM. The primary clearance pathway for AE90015 is mono-oxidation, accounting for 96% of the total, while direct excretion via renal and bile routes accounted for only 0.5%. Bile emerged as the predominant excretion route, at 65%, for metabolites and a minor amount of parent compound, which contrasts with the common assumption that urine would be the primary excretion pathway, which accounted for 26%. Each adamantyl and pyrazine moiety of AE90015 undergoes a one-time oxidation, while the pyridyl portion remains unmetabolized. Secondary metabolites, such as di-hydroxylated forms and glucuronide conjugates, do not contribute to clearance. In this work, a new quantification method combining UV and mass spectra integration was developed, allowing for the quantification of overlapping metabolite peaks. This novel approach proved to be highly effective for metabolite identification in early preclinical studies.
Collapse
Affiliation(s)
- Zhiyang Zack Zou
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Ming-Jie Han
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Yu Chang
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Guiying Li
- TB Alliance, 80 Pine St. 20th Floor, New York, NY 10005, USA
| |
Collapse
|
3
|
Kochoian BA, Bure C, Papa SM. Targeting Striatal Glutamate and Phosphodiesterases to Control L-DOPA-Induced Dyskinesia. Cells 2023; 12:2754. [PMID: 38067182 PMCID: PMC10706484 DOI: 10.3390/cells12232754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
A large body of work during the past several decades has been focused on therapeutic strategies to control L-DOPA-induced dyskinesias (LIDs), common motor complications of long-term L-DOPA therapy in Parkinson's disease (PD). Yet, LIDs remain a clinical challenge for the management of patients with advanced disease. Glutamatergic dysregulation of striatal projection neurons (SPNs) appears to be a key contributor to altered motor responses to L-DOPA. Targeting striatal hyperactivity at the glutamatergic neurotransmission level led to significant preclinical and clinical trials of a variety of antiglutamatergic agents. In fact, the only FDA-approved treatment for LIDs is amantadine, a drug with NMDAR antagonistic actions. Still, novel agents with improved pharmacological profiles are needed for LID therapy. Recently other therapeutic targets to reduce dysregulated SPN activity at the signal transduction level have emerged. In particular, mechanisms regulating the levels of cyclic nucleotides play a major role in the transduction of dopamine signals in SPNs. The phosphodiesterases (PDEs), a large family of enzymes that degrade cyclic nucleotides in a specific manner, are of special interest. We will review the research for antiglutamatergic and PDE inhibition strategies in view of the future development of novel LID therapies.
Collapse
Affiliation(s)
- Brik A. Kochoian
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Cassandra Bure
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Stella M. Papa
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30329, USA
| |
Collapse
|
4
|
Wells-Gatnik WD, Wences Chirino TY, Onan FN, Onan D, Martelletti P. Emerging experimental drugs in clinical trials for migraine: observations and key talking points. Expert Opin Investig Drugs 2023; 32:761-771. [PMID: 37672405 DOI: 10.1080/13543784.2023.2254691] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION There have been significant advances in the treatment of migraine. In response to the clinical success of monoclonal antibodies targeting calcitonin gene-related peptide, there is interest in the clinical trial outcomes of alternative emerging drugs that act on novel targets associated with migraine pathophysiology. As approximately 50% of patients do not respond to CGRP therapies, there is significant value in future drug innovation. Emerging drugs in clinical trials for the treatment of migraine aim to fill this need. AREAS COVERED The emerging drugs that will be discussed in this review include zavegepant, lasmiditan, delta opioid receptor agonists, neuronal nitric oxide synthase inhibitors, monoclonal antibodies targeting pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor, dual orexin receptor antagonists, metabotropic glutamate receptor 5 antagonists, and inducers of ketosis. EXPERT OPINION When considering the preclinical and clinical research related to the emerging drug classes discussed in this review, most therapies are derived from highly supported targets of migraine pathogenesis. Although the individual drugs discussed in this review may be of dubious clinical value, the importance of the therapeutic targets on which they act cannot be understated. Future research is necessary to appropriately target the pathways elucidated by preclinical studies.
Collapse
Affiliation(s)
| | | | | | - Dilara Onan
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Türkiye
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
5
|
Role of P11 through serotonergic and glutamatergic pathways in LID. Mol Biol Rep 2023; 50:4535-4549. [PMID: 36853472 DOI: 10.1007/s11033-023-08326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 02/09/2023] [Indexed: 03/01/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder caused by the degeneration of dopaminergic neurons. This leads to the pathogenesis of multiple basal ganglia-thalamomotor loops and diverse neurotransmission alterations. Dopamine replacement therapy, and on top of that, levodopa and l-3,4-dihydroxyphenylalanine (L-DOPA), is the gold standard treatment, while it develops numerous complications. Levodopa-induced dyskinesia (LID) is well-known as the most prominent side effect. Several studies have been devoted to tackling this problem. Studies showed that metabotropic glutamate receptor 5 (mGluR5) antagonists and 5-hydroxytryptamine receptor 1B (5HT1B) agonists significantly reduced LID when considering the glutamatergic overactivity and compensatory mechanisms of serotonergic neurons after L-DOPA therapy. Moreover, it is documented that these receptors act through an adaptor protein called P11 (S100A10). This protein has been thought to play a crucial role in LID due to its interactions with numerous ion channels and receptors. Lately, experiments have shown successful evidence of the effects of P11 blockade on alleviating LID greater than 5HT1B and mGluR5 manipulations. In contrast, there is a trace of ambiguity in the exact mechanism of action. P11 has shown the potential to be a promising target to diminish LID and prolong L-DOPA therapy in parkinsonian patients owing to further studies and experiments.
Collapse
|
6
|
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
|
7
|
Frouni I, Huot P. Glutamate modulation for the treatment of levodopa induced dyskinesia: a brief review of the drugs tested in the clinic. Neurodegener Dis Manag 2022; 12:203-214. [PMID: 35587024 DOI: 10.2217/nmt-2021-0055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Levodopa is the standard treatment for Parkinson's disease, but its use is marred by the emergence of dyskinesia, for which treatment options remain limited. Here, we review the glutamatergic modulators that were assessed for their antidyskinetic potential in clinical trials, including N-methyl-D-aspartate (NMDA) antagonists, agonists at the glycine-binding site on NMDA receptors, metabotropic glutamate (mGlu) 4 agonists, mGlu5 antagonists, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonists and glutamate release inhibitors. Several agents that were investigated are not selective for their targets, raising uncertainty about the extent to which glutamatergic modulation contributed to their effects. Except for amantadine, the use of glutamatergic modulators for the treatment of dyskinesia in Parkinson's disease remains largely investigational, with promising results obtained with mGlu5 negative allosteric modulation.
Collapse
Affiliation(s)
- Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada.,Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Neuroscience, Division of Neurology, McGill University Health Centre, Montreal, QC, H3A 2B4, Canada
| |
Collapse
|
8
|
Azam S, Jakaria M, Kim J, Ahn J, Kim IS, Choi DK. Group I mGluRs in Therapy and Diagnosis of Parkinson’s Disease: Focus on mGluR5 Subtype. Biomedicines 2022; 10:biomedicines10040864. [PMID: 35453614 PMCID: PMC9032558 DOI: 10.3390/biomedicines10040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs; members of class C G-protein-coupled receptors) have been shown to modulate excitatory neurotransmission, regulate presynaptic extracellular glutamate levels, and modulate postsynaptic ion channels on dendritic spines. mGluRs were found to activate myriad signalling pathways to regulate synapse formation, long-term potentiation, autophagy, apoptosis, necroptosis, and pro-inflammatory cytokines release. A notorious expression pattern of mGluRs has been evident in several neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and schizophrenia. Among the several mGluRs, mGluR5 is one of the most investigated types of considered prospective therapeutic targets and potential diagnostic tools in neurodegenerative diseases and neuropsychiatric disorders. Recent research showed mGluR5 radioligands could be a potential tool to assess neurodegenerative disease progression and trace respective drugs’ kinetic properties. This article provides insight into the group I mGluRs, specifically mGluR5, in the progression and possible therapy for PD.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Md. Jakaria
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - JoonSoo Kim
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Jaeyong Ahn
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
9
|
Striatal glutamatergic hyperactivity in Parkinson's disease. Neurobiol Dis 2022; 168:105697. [DOI: 10.1016/j.nbd.2022.105697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
|
10
|
McCullock TW, Kammermeier PJ. The evidence for and consequences of metabotropic glutamate receptor heterodimerization. Neuropharmacology 2021; 199:108801. [PMID: 34547332 DOI: 10.1016/j.neuropharm.2021.108801] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) are an essential component of the mammalian central nervous system. These receptors modulate neuronal excitability in response to extracellular glutamate through the activation of intracellular heterotrimeric G proteins. Like most other class C G protein-coupled receptors, mGluRs function as obligate dimer proteins, meaning they need to form dimer complexes before becoming functional receptors. All mGluRs possess the ability to homodimerize, but studies over the past ten years have demonstrated these receptors are also capable of forming heterodimers in specific patterns. These mGluR heterodimers appear to have their own unique biophysical behavior and pharmacology with both native and synthetic compounds with few rules having been identified that allow for prediction of the consequences of any particular mGluR pair forming heterodimers. Here, we review the relevant literature demonstrating the existence and consequences of mGluR heterodimerization. By collecting biophysical and pharmacological data of several mGluR heterodimers we demonstrate the lack of generalizable behavior of these complexes indicating that each individual dimeric pair needs to be investigated independently. Additionally, by combining sequence alignment and structural analysis, we propose that interactions between the β4-A Helix Loop and the D Helix in the extracellular domain of these receptors are the structural components that dictate heterodimerization compatibility. Finally, we discuss the potential implications of mGluR heterodimerization from the viewpoints of further developing our understanding of neuronal physiology and leveraging mGluRs as a therapeutic target for the treatment of pathophysiology.
Collapse
Affiliation(s)
- Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| |
Collapse
|
11
|
Gonzalez-Lozano MA, Wortel J, van der Loo RJ, van Weering JRT, Smit AB, Li KW. Reduced mGluR5 Activity Modulates Mitochondrial Function. Cells 2021; 10:cells10061375. [PMID: 34199502 PMCID: PMC8228325 DOI: 10.3390/cells10061375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is an essential modulator of synaptic plasticity, learning and memory; whereas in pathological conditions, it is an acknowledged therapeutic target that has been implicated in multiple brain disorders. Despite robust pre-clinical data, mGluR5 antagonists failed in several clinical trials, highlighting the need for a better understanding of the mechanisms underlying mGluR5 function. In this study, we dissected the molecular synaptic modulation mediated by mGluR5 using genetic and pharmacological mouse models to chronically and acutely reduce mGluR5 activity. We found that next to dysregulation of synaptic proteins, the major regulation in protein expression in both models concerned specific processes in mitochondria, such as oxidative phosphorylation. Second, we observed morphological alterations in shape and area of specifically postsynaptic mitochondria in mGluR5 KO synapses using electron microscopy. Third, computational and biochemical assays suggested an increase of mitochondrial function in neurons, with increased level of NADP/H and oxidative damage in mGluR5 KO. Altogether, our observations provide diverse lines of evidence of the modulation of synaptic mitochondrial function by mGluR5. This connection suggests a role for mGluR5 as a mediator between synaptic activity and mitochondrial function, a finding which might be relevant for the improvement of the clinical potential of mGluR5.
Collapse
Affiliation(s)
- Miguel A. Gonzalez-Lozano
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
- Correspondence: (M.A.G.-L.); (K.W.L.)
| | - Joke Wortel
- Center for Neurogenomics and Cognitive Research, Department of Functional Genomics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (J.W.); (J.R.T.v.W.)
| | - Rolinka J. van der Loo
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
| | - Jan R. T. van Weering
- Center for Neurogenomics and Cognitive Research, Department of Functional Genomics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (J.W.); (J.R.T.v.W.)
- Center for Neurogenomics and Cognitive Research, Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1081 Amsterdam, The Netherlands
| | - August B. Smit
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
| | - Ka Wan Li
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
- Correspondence: (M.A.G.-L.); (K.W.L.)
| |
Collapse
|
12
|
Pivotal Role of Fyn Kinase in Parkinson's Disease and Levodopa-Induced Dyskinesia: a Novel Therapeutic Target? Mol Neurobiol 2020; 58:1372-1391. [PMID: 33175322 DOI: 10.1007/s12035-020-02201-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
The exact etiology of Parkinson's disease (PD) remains obscure, although many cellular mechanisms including α-synuclein aggregation, oxidative damage, excessive neuroinflammation, and dopaminergic neuronal apoptosis are implicated in its pathogenesis. There is still no disease-modifying treatment for PD and the gold standard therapy, chronic use of levodopa is usually accompanied by severe side effects, mainly levodopa-induced dyskinesia (LID). Hence, the elucidation of the precise underlying molecular mechanisms is of paramount importance. Fyn is a tyrosine phospho-transferase of the Src family nonreceptor kinases that is highly implicated in immune regulation, cell proliferation and normal brain development. Accumulating preclinical evidence highlights the emerging role of Fyn in key aspects of PD and LID pathogenesis: it may regulate α-synuclein phosphorylation, oxidative stress-induced dopaminergic neuronal death, enhanced neuroinflammation and glutamate excitotoxicity by mediating key signaling pathways, such as BDNF/TrkB, PKCδ, MAPK, AMPK, NF-κB, Nrf2, and NMDAR axes. These findings suggest that therapeutic targeting of Fyn or Fyn-related pathways may represent a novel approach in PD treatment. Saracatinib, a nonselective Fyn inhibitor, has already been tested in clinical trials for Alzheimer's disease, and novel selective Fyn inhibitors are under investigation. In this comprehensive review, we discuss recent evidence on the role of Fyn in the pathogenesis of PD and LID and provide insights on additional Fyn-related molecular mechanisms to be explored in PD and LID pathology that could aid in the development of future Fyn-targeted therapeutic approaches.
Collapse
|
13
|
Waters S, Sonesson C, Svensson P, Tedroff J, Carta M, Ljung E, Gunnergren J, Edling M, Svanberg B, Fagerberg A, Kullingsjö J, Hjorth S, Waters N. Preclinical Pharmacology of [2-(3-Fluoro-5-Methanesulfonyl-phenoxy)Ethyl](Propyl)amine (IRL790), a Novel Dopamine Transmission Modulator for the Treatment of Motor and Psychiatric Complications in Parkinson Disease. J Pharmacol Exp Ther 2020; 374:113-125. [PMID: 32358046 DOI: 10.1124/jpet.119.264226] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/02/2020] [Indexed: 12/23/2022] Open
Abstract
IRL790 ([2-(3-fluoro-5-methanesulfonylphenoxy)ethyl](propyl)amine, mesdopetam) is a novel compound in development for the clinical management of motor and psychiatric disabilities in Parkinson disease. The discovery of IRL790 was made applying a systems pharmacology approach based on in vivo response profiling. The chemical design idea was to develop a new type of DA D3/D2 receptor type antagonist built on agonist rather than antagonist structural motifs. We hypothesized that such a dopamine antagonist with physicochemical properties similar to agonists would exert antidyskinetic and antipsychotic effects in states of dysregulated dopaminergic signaling while having little negative impact on physiologic dopamine transmission and, hence, minimal liability for side effects related to dopamine-dependent functions. At the level of in vivo pharmacology, IRL790 displays balancing effects on aberrant motor phenotypes, reducing l-DOPA-induced dyskinesias in the rodent 6-hydroxydopamine lesion model and reducing psychostimulant-induced locomotor hyperactivity elicited by pretreatment with either d-amphetamine or dizocilpine, without negatively impacting normal motor performance. Thus, IRL790 has the ability to normalize the behavioral phenotype in hyperdopaminergic as well as hypoglutamatergic states. Neurochemical and immediate early gene (IEG) response profiles suggest modulation of DA neurotransmission, with some features, such as increased DA metabolites and extracellular DA, shared by atypical antipsychotics and others, such as increased frontal cortex IEGs, unique to IRL790. IRL790 also increases extracellular levels of acetylcholine in the prefrontal cortex and ventral hippocampus. At the receptor level, IRL790 appears to act as a preferential DA D3 receptor antagonist. Computational docking studies support preferential affinity at D3 receptors with an agonist-like binding mode. SIGNIFICANCE STATEMENT: This paper reports preclinical pharmacology along with molecular modeling results on IRL790, a novel compound in clinical development for the treatment of motor and psychiatric complications in advanced Parkinson disease. IRL790 is active in models of perturbed dopaminergic and glutamatergic signaling, including rodent 6-hydroxydopamine l-DOPA-induced dyskinesias and psychostimulant-induced hyperactivity, in a dose range that does not impair normal behavior. This effect profile is attributed to interactions at dopamine D2/D3 receptors, with a 6- to 8-fold preference for the D3 subtype.
Collapse
Affiliation(s)
- Susanna Waters
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Clas Sonesson
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Peder Svensson
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Joakim Tedroff
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Manolo Carta
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Elisabeth Ljung
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Jenny Gunnergren
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Malin Edling
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Boel Svanberg
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Anne Fagerberg
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Johan Kullingsjö
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Stephan Hjorth
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Nicholas Waters
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| |
Collapse
|
14
|
Crabbé M, Dirkx N, Casteels C, Laere KV. Excitotoxic neurodegeneration is associated with a focal decrease in metabotropic glutamate receptor type 5 availability: an in vivo PET imaging study. Sci Rep 2019; 9:12916. [PMID: 31501497 PMCID: PMC6733799 DOI: 10.1038/s41598-019-49356-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/23/2019] [Indexed: 11/09/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) have been proposed as promising therapeutic targets to correct the dysregulated glutamate signaling, associated with neurodegenerative pathologies. Of all mGluR subtypes, especially mGluR5 acts as a modulator of glutamate-induced excitotoxicity. To study the behavior of mGluR5 following localized excitotoxicity, we utilised a pharmacological model that portrays exacerbated neuronal glutamate release, mediated by the endogenous excitotoxin quinolinic acid (QA). Using longitudinal positron emission tomography (PET) with [18F]FPEB, we investigated cerebral changes in mGluR5 following striatal QA-lesioning. Behavioral tests were executed to monitor motor and cognitive performance. Decreased mGluR5 binding potential (BPND) was found in the affected striatum and globus pallidus of QA-lesioned rats at week 3, and further decreased at week 7, as compared to sham-injected controls. mGluR5 availability in the ipsilateral nucleus accumbens was significantly decreased at 7 weeks post-injection. QA rats performed significantly worse on motor coordination and balance compared to control rats. Correlation analysis indicated a positive correlation between striatal mGluR5 BPND and rotarod performance whereas print width of the unaffected forepaws showed a positive relation with mGluR5 BPND in the contralateral motor cortex. Together, our results suggest decreased mGluR5 availability to be related to excitotoxin-induced neurodegeneration and symptomatology although late stage effects do indicate possible cortical mGluR5-mediated effects on motor behavior.
Collapse
Affiliation(s)
- Melissa Crabbé
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium. .,MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium.
| | - Nina Dirkx
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium.,MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium
| | - Cindy Casteels
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium.,MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium.,MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Masilamoni GJ, Smith Y. Group I metabotropic glutamate receptors in the primate motor thalamus: subsynaptic association with cortical and sub-cortical glutamatergic afferents. Brain Struct Funct 2019; 224:2787-2804. [PMID: 31422483 DOI: 10.1007/s00429-019-01937-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
Preclinical evidence indicates that mGluR5 is a potential therapeutic target for Parkinson's disease and L-DOPA-induced dyskinesia. However, the mechanisms through which these therapeutic benefits are mediated remain poorly understood. Although the regulatory role of mGluR5 on glutamatergic transmission has been examined in various basal ganglia nuclei, very little is known about the localization and function of mGluR5 in the ventral motor and intralaminar thalamic nuclei, the main targets of basal ganglia output in mammals. Thus, we used immuno-electron microscopy to map the cellular and subcellular localization of group I mGluRs (mGluR1a and mGluR5) in the ventral motor and caudal intralaminar thalamic nuclei in rhesus monkeys. Furthermore, using double immuno-electron microscopy, we examined the subsynaptic localization of mGluR5 in relation to cortical and sub-cortical glutamatergic afferents. Four major conclusions can be drawn from these data. First, mGluR1a and mGluR5 are expressed postsynaptically on the plasma membrane of dendrites of projection neurons and GABAergic interneurons in the basal ganglia- and cerebellar-receiving regions of the ventral motor thalamus and in CM. Second, the plasma membrane-bound mGluR5 immunoreactivity is preferentially expressed perisynaptically at the edges of cortical and sub-cortical glutamatergic afferents. Third, the mGluR5 immunoreactivity is more strongly expressed in the lateral than the medial tiers of CM, suggesting a preferential association with thalamocortical over thalamostriatal neurons in the primate CM. Overall, mGluR5 is located to subserve powerful modulatory role of cortical and subcortical glutamatergic transmission in the primate ventral motor thalamus and CM.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA. .,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
16
|
Pourmirbabaei S, Dolatshahi M, Rahmani F. Pathophysiological clues to therapeutic applications of glutamate mGlu5 receptor antagonists in levodopa-induced dyskinesia. Eur J Pharmacol 2019; 855:149-159. [PMID: 31063776 DOI: 10.1016/j.ejphar.2019.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/16/2022]
Abstract
Levodopa remains to be the mainstay for treatment of Parkinson disease (PD). Long-term levodopa treatment bears a risk for developing levodopa-induced dyskinesia (LID). LID significantly overshadows patients' quality of life and therapeutic efficacy of levodopa. Pre- and post-synaptic changes in dopamine secretion and signaling, along with altered glutamate receptor expression and glutamatergic signaling in striatal neurons, and the resulting disinhibition-like changes in the corticostriatal circuitry, lead to aberrant activity of motor cortex and formation of LID. Research has highlighted the role of group I metabotropic glutamate receptors especially the metabotropic glutamate receptor 5 (mGlu5) in formation of LID through potentiating of ionotropic glutamate NMDA receptors and dopamine D1/D5 receptors in direct pathway. Accordingly, MTEP and MPEP were the first mGlu5 receptor antagonists which were shown to attenuate LID in animal models through suppression of downstream signaling cascades involving mitogen-activated protein kinase (MAPK) and FosB/delta FosB activation, as well as modulation of prodynorphinegic, preproenkephalinergic, and GABA-ergic neurotransmission systems. Beneficial effects of other mGlu5 receptor antagonists such as AFQ056/mavoglurant and ADX48621/dipraglurant in amelioration of LID has been shown not only in animal models but also in clinical trials. Considering the presence of mGlu receptor dysregulation in rapid eye movement (REM) sleep behavior disorder and depression, which are prodromal signs of PD, along with the neuroprotective effects of mGlu receptor antagonists, and their cognitive benefits, potential effectiveness of mGlu receptor antagonists in early prevention of PD remains to be investigated.
Collapse
Affiliation(s)
- Shayan Pourmirbabaei
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Dolatshahi
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Rahmani
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|