1
|
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
|
2
|
Mao LM, Mathur N, Shah K, Wang JQ. Roles of metabotropic glutamate receptor 8 in neuropsychiatric and neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:349-366. [PMID: 36868634 PMCID: PMC10162486 DOI: 10.1016/bs.irn.2022.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are G protein-coupled receptors. Among eight mGlu subtypes (mGlu1-8), mGlu8 has drawn increasing attention. This subtype is localized to the presynaptic active zone of neurotransmitter release and is among the mGlu subtypes with high affinity for glutamate. As a Gi/o-coupled autoreceptor, mGlu8 inhibits glutamate release to maintain homeostasis of glutamatergic transmission. mGlu8 receptors are expressed in limbic brain regions and play a pivotal role in modulating motivation, emotion, cognition, and motor functions. Emerging evidence emphasizes the increasing clinical relevance of abnormal mGlu8 activity. Studies using mGlu8 selective agents and knockout mice have revealed the linkage of mGlu8 receptors to multiple neuropsychiatric and neurological disorders, including anxiety, epilepsy, Parkinson's disease, drug addiction, and chronic pain. Expression and function of mGlu8 receptors in some limbic structures undergo long-lasting adaptive changes in animal models of these disorders, which may contribute to the remodeling of glutamatergic transmission critical for the pathogenesis and symptomatology of brain illnesses. This review summarizes the current understanding of mGlu8 biology and the possible involvement of the receptor in several common psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - Nirav Mathur
- Department of Anesthesiology, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - Karina Shah
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - John Q Wang
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States; Department of Anesthesiology, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States.
| |
Collapse
|
3
|
Rai SN, Singh P, Varshney R, Chaturvedi VK, Vamanu E, Singh MP, Singh BK. Promising drug targets and associated therapeutic interventions in Parkinson's disease. Neural Regen Res 2021; 16:1730-1739. [PMID: 33510062 PMCID: PMC8328771 DOI: 10.4103/1673-5374.306066] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is one of the most debilitating brain diseases. Despite the availability of symptomatic treatments, response towards the health of PD patients remains scarce. To fulfil the medical needs of the PD patients, an efficacious and etiological treatment is required. In this review, we have compiled the information covering limitations of current therapeutic options in PD, novel drug targets for PD, and finally, the role of some critical beneficial natural products to control the progression of PD.
Collapse
Affiliation(s)
| | - Payal Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ritu Varshney
- Department of Bioengineering and Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | | | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, Bucharest, Romania
| | - M. P. Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj, India
| | - Brijesh Kumar Singh
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Mann E, Jackson M, Lincoln L, Fisher R, Rose S, Duty S. Antiparkinsonian Effects of a Metabotropic Glutamate Receptor 4 Agonist in MPTP-Treated Marmosets. JOURNAL OF PARKINSONS DISEASE 2021; 10:959-967. [PMID: 32250315 DOI: 10.3233/jpd-191824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increased firing across glutamatergic synapses may contribute to both the motor dysfunction and L-DOPA-induced dyskinesia seen in Parkinson's disease. Given their ability to reduce glutamate release, activation of group III metabotropic glutamate receptors such as metabotropic glutamate receptor 4 may prove effective against both motor dysfunction and dyskinesia in Parkinson's disease. OBJECTIVE We hypothesised that activation of metabotropic glutamate receptor 4 by an orthosteric agonist ((2S)-2-amino-4-(hydroxy(hydroxy(4-hydroxy-3-methoxy-5-nitrophenyl)methyl)phosphoryl)butanoic acid, LSP1-2111) would produce antiparkinsonian activity and reduce expression of dyskinesia in a 1-methyl-4-phenyl,1,2,3,6-tetrahydropyridine (MPTP)-treated marmoset model of Parkinson's disease. METHODS Common marmosets were previously treated with MPTP and pre-primed with L-DOPA for up to 28 days to express dyskinesia. LSP1-2111 (1, 3, or 6 mg/kg s.c.) or vehicle (0.9% saline s.c.) were administered immediately prior to L-DOPA (8 mg/kg + benserazide (10 mg/kg) p.o.) or vehicle (10% sucrose p.o.). Locomotor activity was measured in automated test cages and animals were scored for dyskinesia and disability. RESULTS As expected, L-DOPA reversed motor disability and induced moderate dyskinesia. By contrast, LSP1-2111 alone significantly reduced the motor disability without any accompanying expression of dyskinesia. When administered in combination with L-DOPA, LSP1-2111 did not significantly reduce the severity of L-DOPA-induced dyskinesia. CONCLUSION Systemic administration of LSP1-2111 reduces motor disability without causing dyskinesia in MPTP-treated marmosets, supporting a role for metabotropic glutamate receptor 4 orthosteric agonists as promising monotherapy for PD. Conversely, this study found no evidence to support their use as antidyskinetic agents within the dose range tested.
Collapse
Affiliation(s)
- Elizabeth Mann
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Michael Jackson
- Neurodegenerative Diseases Research Group, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Louise Lincoln
- Neurodegenerative Diseases Research Group, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Ria Fisher
- Neurodegenerative Diseases Research Group, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Sarah Rose
- Neurodegenerative Diseases Research Group, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Susan Duty
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
5
|
Behl T, Kaur I, Sehgal A, Singh S, Bhatia S, Al-Harrasi A, Zengin G, Bumbu AG, Andronie-Cioara FL, Nechifor AC, Gitea D, Bungau AF, Toma MM, Bungau SG. The Footprint of Kynurenine Pathway in Neurodegeneration: Janus-Faced Role in Parkinson's Disorder and Therapeutic Implications. Int J Mol Sci 2021; 22:6737. [PMID: 34201647 PMCID: PMC8268239 DOI: 10.3390/ijms22136737] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Progressive degeneration of neurons and aggravation of dopaminergic neurons in the substantia nigra pars compacta results in the loss of dopamine in the brain of Parkinson's disease (PD) patients. Numerous therapies, exhibiting transient efficacy have been developed; however, they are mostly accompanied by side effects and limited reliability, therefore instigating the need to develop novel optimistic treatment targets. Significant therapeutic targets have been identified, namely: chaperones, protein Abelson, glucocerebrosidase-1, calcium, neuromelanin, ubiquitin-proteasome system, neuroinflammation, mitochondrial dysfunction, and the kynurenine pathway (KP). The role of KP and its metabolites and enzymes in PD, namely quinolinic acid (QUIN), kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK), 3-hydroxyanthranillic acid (3-HAA), kunurenine-3-monooxygenase (KMO), etc. has been reported. The neurotoxic QUIN, N-methyl-D-aspartate (NMDA) receptor agonist, and neuroprotective KYNA-which antagonizes QUIN actions-primarily justify the Janus-faced role of KP in PD. Moreover, KP has been reported to play a biomarker role in PD detection. Therefore, the authors detail the neurotoxic, neuroprotective, and immunomodulatory neuroactive components, alongside the upstream and downstream metabolic pathways of KP, forming a basis for a therapeutic paradigm of the disease while recognizing KP as a potential biomarker in PD, thus facilitating the development of a suitable target in PD management.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Gurugram, Haryana 122412, India;
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616 Birkat Al Mouz, Nizwa 611, Oman;
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616 Birkat Al Mouz, Nizwa 611, Oman;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42130, Turkey;
| | - Adrian Gheorghe Bumbu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Aurelia Cristina Nechifor
- Analytical Chemistry and Environmental Engineering Department, Polytechnic University of Bucharest, 011061 Bucharest, Romania;
| | - Daniela Gitea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania; (D.G.); (M.M.T.)
| | | | - Mirela Marioara Toma
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania; (D.G.); (M.M.T.)
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania; (D.G.); (M.M.T.)
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
6
|
Kobiec T, Otero-Losada M, Chevalier G, Udovin L, Bordet S, Menéndez-Maissonave C, Capani F, Pérez-Lloret S. The Renin-Angiotensin System Modulates Dopaminergic Neurotransmission: A New Player on the Scene. Front Synaptic Neurosci 2021; 13:638519. [PMID: 33967734 PMCID: PMC8100578 DOI: 10.3389/fnsyn.2021.638519] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is an extrapyramidal disorder characterized by neuronal degeneration in several regions of the peripheral and central nervous systems. It is the second most frequent neurodegenerative disease after Alzheimer's. It has become a major health problem, affecting 1% of the world population over 60 years old and 3% of people beyond 80 years. The main histological findings are intracellular Lewy bodies composed of misfolded α-synuclein protein aggregates and loss of dopaminergic neurons in the central nervous system. Neuroinflammation, apoptosis, mitochondrial dysfunction, altered calcium homeostasis, abnormal protein degradation, and synaptic pathobiology have been put forward as mechanisms leading to cell death, α-synuclein deposition, or both. A progressive loss of dopaminergic neurons in the substantia nigra late in the neurodegeneration leads to developing motor symptoms like bradykinesia, tremor, and rigidity. The renin-angiotensin system (RAS), which is involved in regulating blood pressure and body fluid balance, also plays other important functions in the brain. The RAS is involved in the autocrine and paracrine regulation of the nigrostriatal dopaminergic synapses. Dopamine depletion, as in PD, increases angiotensin II expression, which stimulates or inhibits dopamine synthesis and is released via AT1 or AT2 receptors. Furthermore, angiotensin II AT1 receptors inhibit D1 receptor activation allosterically. Therefore, the RAS may have an important modulating role in the flow of information from the brain cortex to the basal ganglia. High angiotensin II levels might even aggravate neurodegeneration, activating the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, which leads to increased reactive oxygen species production.
Collapse
Affiliation(s)
- Tamara Kobiec
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Guenson Chevalier
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lucas Udovin
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Sofía Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Camila Menéndez-Maissonave
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
- Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Biología, Universidad Argentina John F. Kennedy, Buenos Aires, Argentina
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago de Chile, Chile
| | - Santiago Pérez-Lloret
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Medicina, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
O'Rourke T, Martins PT, Asano R, Tachibana RO, Okanoya K, Boeckx C. Capturing the Effects of Domestication on Vocal Learning Complexity. Trends Cogn Sci 2021; 25:462-474. [PMID: 33810982 DOI: 10.1016/j.tics.2021.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 11/28/2022]
Abstract
Domesticated and vocal learning species can serve as informative model organisms for the reduction of reactive aggression and emergence of speech in our lineage. Amidst mounting evidence that domestication modifies vocal repertoires across different species, we focus on the domesticated Bengalese finch, which has a more complex song than the wild-type white-rumped munia. Our explanation for this effect revolves around the glutamate neurotransmitter system. Glutamate signaling (i) is implicated in birdsong learning, (ii) controls dopamine activity in neural circuits crucial for vocal learning, (iii) is disproportionately targeted in the evolution of domesticates, and (iv) regulates stress responses and aggressive behaviors attenuated under domestication. We propose that attenuated excitation of stress-related neural circuits potentiates vocal learning via altered dopaminergic signaling.
Collapse
Affiliation(s)
- Thomas O'Rourke
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain
| | - Pedro Tiago Martins
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain
| | - Rie Asano
- Department of Systematic Musicology, University of Cologne, 50923 Cologne, Germany
| | - Ryosuke O Tachibana
- Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Tokyo, Japan
| | - Kazuo Okanoya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Tokyo, Japan
| | - Cedric Boeckx
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain; Catalan Institute for Advanced Studies and Research (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
8
|
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
|
9
|
Charvin D, Di Paolo T, Bezard E, Gregoire L, Takano A, Duvey G, Pioli E, Halldin C, Medori R, Conquet F. An mGlu4-Positive Allosteric Modulator Alleviates Parkinsonism in Primates. Mov Disord 2018; 33:1619-1631. [PMID: 30216534 DOI: 10.1002/mds.27462] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 04/19/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Levodopa remains the gold-standard treatment for PD. However, it becomes less effective as the disease progresses and produces debilitating side effects, such as motor fluctuations and l-dopa-induced dyskinesia. Modulation of metabotropic glutamate receptor 4 represents a promising antiparkinsonian approach in combination with l-dopa, but it has not been demonstrated in primates. OBJECTIVE We studied whether a novel positive allosteric modulator of the metabotropic glutamate receptor 4, PXT002331 (foliglurax), could reduce parkinsonism in primate models. METHODS We assessed the therapeutic potential of PXT002331 in three models of MPTP-induced parkinsonism in macaques. These models represent three different stages of disease evolution: early stage and advanced stage with and without l-dopa-induced dyskinesia. RESULTS As an adjunct to l-dopa, PXT002331 induced a robust and dose-dependent reversal of parkinsonian motor symptoms in macaques, including bradykinesia, tremor, posture, and mobility. Moreover, PXT002331 strongly decreased dyskinesia severity, thus having therapeutic efficacy on both parkinsonian motor impairment and l-dopa-induced dyskinesia. PXT002331 brain penetration was also assessed using PET imaging in macaques, and pharmacodynamic analyses support target engagement in the therapeutic effects of PXT002331. CONCLUSIONS This work provides a demonstration that a positive allosteric modulator of metabotropic glutamate receptor 4 can alleviate the motor symptoms of PD and the motor complications induced by l-dopa in primates. PXT002331 is the first compound of its class to enter phase IIa clinical trials. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Delphine Charvin
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Therese Di Paolo
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Erwan Bezard
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Laurent Gregoire
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Akihiro Takano
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Guillaume Duvey
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Elsa Pioli
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Christer Halldin
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Rossella Medori
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - François Conquet
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| |
Collapse
|
10
|
Du JJ, Chen SD. Current Nondopaminergic Therapeutic Options for Motor Symptoms of Parkinson's Disease. Chin Med J (Engl) 2018; 130:1856-1866. [PMID: 28748860 PMCID: PMC5547839 DOI: 10.4103/0366-6999.211555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: The aim of this study was to summarize recent studies on nondopaminergic options for the treatment of motor symptoms in Parkinson's disease (PD). Data Sources: Papers in English published in PubMed, Cochrane, and Ovid Nursing databases between January 1988 and November 2016 were searched using the following keywords: PD, nondopaminergic therapy, adenosine, glutamatergic, adrenergic, serotoninergic, histaminic, and iron chelator. We also reviewed the ongoing clinical trials in the website of clinicaltrials.gov. Study Selection: Articles related to the nondopaminergic treatment of motor symptoms in PD were selected for this review. Results: PD is conventionally treated with dopamine replacement strategies, which are effective in the early stages of PD. Long-term use of levodopa could result in motor complications. Recent studies revealed that nondopaminergic systems such as adenosine, glutamatergic, adrenergic, serotoninergic, histaminic, and iron chelator pathways could include potential therapeutic targets for motor symptoms, including motor fluctuations, levodopa-induced dyskinesia, and gait disorders. Some nondopaminergic drugs, such as istradefylline and amantadine, are currently used clinically, while most such drugs are in preclinical testing stages. Transitioning of these agents into clinically beneficial strategies requires reliable evaluation since several agents have failed to show consistent results despite positive findings at the preclinical level. Conclusions: Targeting nondopaminergic transmission could improve some motor symptoms in PD, especially the discomfort of dyskinesia. Although nondopaminergic treatments show great potential in PD treatment as an adjunct therapy to levodopa, further investigation is required to ensure their success.
Collapse
Affiliation(s)
- Juan-Juan Du
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
11
|
Masilamoni GJ, Smith Y. Metabotropic glutamate receptors: targets for neuroprotective therapies in Parkinson disease. Curr Opin Pharmacol 2018; 38:72-80. [PMID: 29605730 DOI: 10.1016/j.coph.2018.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/12/2018] [Indexed: 01/22/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are heavily expressed throughout the basal ganglia (BG), where they modulate neuronal excitability, transmitter release and long term synaptic plasticity. Therefore, targeting specific mGluR subtypes by means of selective drugs could be a possible strategy for restoring normal synaptic function and neuronal activity of the BG in Parkinson disease (PD). Preclinical studies have revealed that specific mGluR subtypes mediate significant neuroprotective effects that reduce toxin-induced midbrain dopaminergic neuronal death in animal models of PD. Although the underlying mechanisms of these effects must be further studied, there is evidence that intracellular calcium regulation, anti-inflammatory effects, and glutamatergic network modulation contribute to some of these neuroprotective properties. It is noteworthy that these protective effects extend beyond midbrain dopaminergic neurons to include other monoaminergic cell groups for some mGluRs. In this review, we discuss evidence for mGluR-mediated neuroprotection in PD and highlight the challenges to translate these findings into human trials.
Collapse
Affiliation(s)
- Gunasingh J Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA.
| |
Collapse
|
12
|
mGlu 4 allosteric modulation for treating Parkinson's disease. Neuropharmacology 2018; 135:308-315. [PMID: 29578036 DOI: 10.1016/j.neuropharm.2018.03.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 12/21/2022]
Abstract
2017 is the 200th anniversary of the first published description of Parkinson's disease (PD). Fifty years ago, the clinical benefit of levodopa was first documented, representing the most important advance in the treatment of PD so far. Among the novel targets identified in the last decade, positive allosteric modulators (PAM) of mGlu4 receptors show great promise, with the potential to change the paradigm of the PD treatment approach. mGlu4 PAMs have shown consistent efficacy in various preclinical models of PD, and entered clinical trials for the first time in 2017. This review synthesizes the rationale for mGlu4 PAM development for PD and progress to date, reporting the key achievements from preclinical studies to the first-in-class compound assessment in man.
Collapse
|
13
|
Yang Q, Zhao C, Zhao J, Ye Y. Synthesis and neuroprotective effects of the complex nanoparticles of iron and sapogenin isolated from the defatted seeds of Camellia oleifera. PHARMACEUTICAL BIOLOGY 2017; 55:428-434. [PMID: 27937035 PMCID: PMC6130765 DOI: 10.1080/13880209.2016.1245346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 05/26/2023]
Abstract
CONTEXT The defatted seeds of Camellia oleifera var. monosperma Hung T. Chang (Theaceae) are currently discarded without effective utilization. However, sapogenin has been isolated and shows antioxidative, anti-inflammatory and analgesic activities suggestive of its neuroprotective function. OBJECTIVE In order to improve the activities of sapogenin, the nanoparticles of iron-sapogenin have been synthesized, and the neuroprotective effects are evaluated. MATERIALS AND METHODS Structural characters of the nanoparticles were analyzed, and the antioxidant effect was assessed by DPPH method, and the neuroprotective effect was evaluated by rotenone-induced neurodegeneration in Kunming mice injected subcutaneously into the back of neck with rotenone (50 mg/kg/day) for 6 weeks and then treated by tail intravenous injection with the iron-sapogenin at the dose of 25, 50 and 100 mg/kg for 7 days. Mice behaviour and neurotransmitters were tested. RESULTS The product had an average size of 162 nm with spherical shape, and scavenged more than 90% DPPH radicals at 0.8 mg/mL concentration. It decreased behavioural disorder and malondialdehyde content in mice brain, and increased superoxide dismutase activity, tyrosine hydroxylase expression, dopamine and acetylcholine levels in brain in dose dependence, and their maximum changes were respectively up to 60.83%, 25.17%, 22.13%, 105.26%, 42.17% and 22.89% as compared to vehicle group. Iron-sapogenin nanoparticle shows significantly better effects than the sapogenin. DISCUSSION AND CONCLUSION Iron-sapogenin alleviates neurodegeneration of mice injured by neurotoxicity of rotenone, it is a superior candidate of drugs for neuroprotection.
Collapse
Affiliation(s)
- Qian Yang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, PR China
| | - Chuang Zhao
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, PR China
| | - Jun Zhao
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, PR China
| | - Yong Ye
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, PR China
| |
Collapse
|
14
|
Involvement of the kynurenine pathway in the pathogenesis of Parkinson’s disease. Prog Neurobiol 2017; 155:76-95. [DOI: 10.1016/j.pneurobio.2015.12.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022]
|
15
|
Jia Y, Deng J, Zhang W, Sun Z, Yang J, Yu Y, Gong X, Jia J, Wang X. The Role of Group II Metabotropic Glutamate Receptors in the Striatum in Electroacupuncture Treatment of Parkinsonian Rats. CNS Neurosci Ther 2017; 23:23-32. [PMID: 27412260 PMCID: PMC6492692 DOI: 10.1111/cns.12587] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 06/11/2016] [Accepted: 06/12/2016] [Indexed: 12/21/2022] Open
Abstract
AIMS Glutamatergic transmission may play a critical role in the pathogenesis of Parkinson's disease (PD). Electroacupuncture (EA) has been demonstrated to effectively alleviate PD symptoms. In this study, a potential glutamate-dependent mechanism underlying the therapeutic action of EA was investigated. METHODS The effects of EA stimulation on motor behaviors, dopamine contents, glutamate release, and group II metabotropic glutamate receptor (mGluR2/3) expression in unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats were examined. RESULTS Unilateral 6-OHDA lesions of the nigrostriatal system caused a marked increase in glutamate content in the ipsilateral cortex and striatum. mGluR2/3 protein expression and mGluR3 mRNA expression were reduced in the striatum. Noticeably, prolonged EA stimulation at 100 Hz significantly reversed these changes in the striatal glutamate system. Behaviorally, EA improved the motor deficits induced by 6-OHDA lesions. Intrastriatal infusion of an mGluR2/3 antagonist APICA blocked the improving effect of EA. CONCLUSIONS These data collectively demonstrate that the group II mGluR-mediated glutamatergic transmission in the striatum is sensitive to dopamine depletion and may serve as a substrate of EA for mediating the therapeutic effect of EA in a rat model of PD.
Collapse
Affiliation(s)
- Yan‐Jun Jia
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jia‐Hui Deng
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Wen‐Zhong Zhang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Zuo‐Li Sun
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jian Yang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Yan Yu
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Xiao‐Li Gong
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jun Jia
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Xiao‐Min Wang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| |
Collapse
|
16
|
The neuroprotective effects of orthosteric agonists of group II and III mGluRs in primary neuronal cell cultures are dependent on developmental stage. Neuropharmacology 2016; 111:195-211. [PMID: 27600687 DOI: 10.1016/j.neuropharm.2016.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/12/2016] [Accepted: 09/02/2016] [Indexed: 11/22/2022]
Abstract
Activation of metabotropic glutamate receptors (mGluRs) modulates neuronal excitability. Here, we evaluated the neuroprotective potential of four structurally diverse activators of group II and III mGluRs: an orthosteric agonist of group II (LY354740), an orthosteric agonist of group III (ACPT-I), an allosteric agonist of mGluR7 (AMN082) and a positive allosteric modulator (PAM) of mGluR4 (VU0361737). Neurotoxicity was induced by the pro-apoptotic agents: staurosporine (St) and doxorubicin (Dox) or the excitotoxic factor glutamate (Glu). The effects were analyzed in primary hippocampal (HIP) and cerebellar granule cell (CGC) cultures at two developmental stages, at 7 and 12 days in vitro (DIV). The data reveal a general neuroprotective effect of group II and III mGluR activators against the St- and Glu- but not Dox-induced cell damage. We found that neuroprotective effects of group II and III mGluR orthosteric agonists (LY354740 and ACPT-I) were higher at 12 DIV when compared to 7 DIV cells. In contrast, the efficiency of allosteric mGluR agents (AMN082 and VU0361737) did not differ between 7 and 12 DIV in both, St and Glu models of neuronal cell damage. Interestingly, the protective effects of activators of group II and III mGluRs were blocked by relevant antagonists only against Glu-induced neurotoxicity. Moreover, the observed neuroprotective action of group II and III mGluR activators in the St model was associated with a decreased number of PI-positive cells and no alterations in the caspase-3 activity. Finally, we showed that MAPK/ERK pathway activation was potentially involved in the mechanism of ACPT-I- and AMN082-induced neuroprotection against the St-evoked cellular damage. Our comparative study demonstrated the developmental stage-dependent neuroprotective effect of orthosteric group II and III mGluR agonists. In comparison to allosteric modulators, orthosteric compounds may provide more specific tools for suppression of neuronal cell loss associated with various chronic neurodegenerative conditions. Our results also suggest that the inhibition of intracellular pathways mediating necrotic, rather than apoptotic cascades, may be involved in neuroprotective effects of activators of group II and III mGluRs.
Collapse
|
17
|
Volta M, Milnerwood AJ, Farrer MJ. Insights from late-onset familial parkinsonism on the pathogenesis of idiopathic Parkinson's disease. Lancet Neurol 2015; 14:1054-64. [PMID: 26376970 DOI: 10.1016/s1474-4422(15)00186-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/24/2023]
Abstract
Disease-modifying therapies that slow or halt the progression of Parkinson's disease are an unmet clinical need. Many hypotheses have been put forward to explain the pathogenesis of the disease, but none has led to the development of disease-modifying drugs. Here we focus on familial forms of late-onset parkinsonism that most closely resemble idiopathic Parkinson's disease and present a synthesis of emerging molecular advances. Genetic discoveries and mechanistic investigations have highlighted early alterations to synaptic function, endosomal maturation, and protein sorting that might lead to an intracellular proteinopathy. We propose that these cellular processes constitute one pathway to pathogenesis and suggest that neuroprotection, as an adjunct to current symptomatic treatments, need not remain an elusive goal.
Collapse
Affiliation(s)
- Mattia Volta
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Austen J Milnerwood
- Division of Neurology, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Ivashkin P, Lemonnier G, Tora AS, Pin JP, Goudet C, Jubault P, Pannecoucke X. Synthesis and studies on the mGluR agonist activity of FAP4 stereoisomers. Bioorg Med Chem Lett 2015; 25:2523-6. [PMID: 25958247 DOI: 10.1016/j.bmcl.2015.04.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 11/20/2022]
Abstract
The four stereoisomers of 1-amino-2-fluoro-2-(phosphonomethyl)cyclopropane-1-carboxylic acid (FAP4) were synthesized via diastereoselective Rh(II)-catalysed cyclopropanation of a phosphonylated fluoroalkene. Different isomers of FAP4 and the corresponding non-fluorinated analogs showed a similar pharmacological profile against the isoforms of metabotropic glutamate receptor (mGluR). Within the fluorinated series, (-)-(Z)-FAP4 and (-)-(E)-FAP4 demonstrated the highest agonist activity against mGlu4 (EC50 0.10 μM). Our results suggest that fluorocyclopropanes bearing an amino-acid function can be suitable for the development of potent conformationally restricted mGluR agonists.
Collapse
Affiliation(s)
- Pavel Ivashkin
- NormandieUniv., COBRA, UMR 6014 & FR 3038, Univ. Rouen, INSA Rouen, CNRS, 1 rue Tesnière, F-76821 Mont-Saint-Aignan Cedex, France
| | - Gérald Lemonnier
- NormandieUniv., COBRA, UMR 6014 & FR 3038, Univ. Rouen, INSA Rouen, CNRS, 1 rue Tesnière, F-76821 Mont-Saint-Aignan Cedex, France
| | - Amélie S Tora
- Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier, F-34094 Montpellier, France; INSERM U1191, F-34094 Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier, F-34094 Montpellier, France; INSERM U1191, F-34094 Montpellier, France
| | - Cyril Goudet
- Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier, F-34094 Montpellier, France; INSERM U1191, F-34094 Montpellier, France
| | - Philippe Jubault
- NormandieUniv., COBRA, UMR 6014 & FR 3038, Univ. Rouen, INSA Rouen, CNRS, 1 rue Tesnière, F-76821 Mont-Saint-Aignan Cedex, France.
| | - Xavier Pannecoucke
- NormandieUniv., COBRA, UMR 6014 & FR 3038, Univ. Rouen, INSA Rouen, CNRS, 1 rue Tesnière, F-76821 Mont-Saint-Aignan Cedex, France
| |
Collapse
|
19
|
Lawrence K, Jackson T, Jamieson D, Stevens A, Owens G, Sayan B, Locke I, Townsend P. Urocortin – From Parkinson's disease to the skeleton. Int J Biochem Cell Biol 2015; 60:130-8. [DOI: 10.1016/j.biocel.2014.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 01/04/2023]
|
20
|
Jantas D, Greda A, Leskiewicz M, Grygier B, Pilc A, Lason W. Neuroprotective effects of mGluR II and III activators against staurosporine- and doxorubicin-induced cellular injury in SH-SY5Y cells: New evidence for a mechanism involving inhibition of AIF translocation. Neurochem Int 2015; 88:124-37. [PMID: 25661514 DOI: 10.1016/j.neuint.2014.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/06/2014] [Accepted: 12/28/2014] [Indexed: 12/12/2022]
Abstract
There are several experimental data sets demonstrating the neuroprotective effects of activation of group II and III metabotropic glutamate receptors (mGluR II/III), however, their effect on neuronal apoptotic processes has yet to be fully recognized. Thus, the comparison of the neuroprotective potency of the mGluR II agonist LY354740, mGluR III agonist ACPT-I, mGluR4 PAM VU0361737, mGluR8 PAM AZ12216052 and allosteric mGluR7 agonist AMN082 against staurosporine (St-) and doxorubicin (Dox)-induced cell death has been performed in undifferentiated (UN-) and retinoic acid differentiated (RA-) human neuroblastoma SH-SY5Y cells. The highest neuroprotection in UN-SH-SY5Y cells was noted for AZ12216052 (0.01-1 µM) and VU0361737 (1-10 µM), with both agents partially attenuating the St- and Dox-evoked cell death. LY354740 (0.01-10 µM) and ACPT-I (10 µM) were protective only against the St-evoked cell damage, whereas AMN082 (0.001-0.01 µM) attenuated only the Dox-induced cell death. In RA-SH-SY5Y, a moderate neuroprotective response of mGluR II/III activators was observed for LY354740 (10 µM) and AZ12216052 (0.01 and 10 µM), which afforded protection only against the St-induced cell damage. The protection mediated by mGluR II/III activators against the St- and Dox-evoked cell death in UN-SH-SY5Y cells was not related to attenuation of caspase-3 activity, however, a decrease in the number of TUNEL-positive nuclei was found. Moreover, mGluR II/III activators attenuated the cytosolic level of the apoptosis inducing factor (AIF), which was increased after St and Dox exposure. Our data point to differential neuroprotective efficacy of various mGluR II/III activators in attenuating St- and Dox-evoked cell damage in SH-SY5Y cells, and dependence of the effects on the cellular differentiation state, as well on the type of the pro-apoptotic agent that is employed. Moreover, the neuroprotection mediated by mGluR II/III activators is accompanied by inhibition of caspase-3-independent DNA fragmentation evoked by AIF translocation.
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland.
| | - A Greda
- Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland
| | - M Leskiewicz
- Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland
| | - B Grygier
- Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland
| | - A Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland
| | - W Lason
- Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna 12 Street, Krakow PL 31-343, Poland
| |
Collapse
|
21
|
Amalric M. Targeting metabotropic glutamate receptors (mGluRs) in Parkinson's disease. Curr Opin Pharmacol 2014; 20:29-34. [PMID: 25462289 DOI: 10.1016/j.coph.2014.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 12/28/2022]
Abstract
The interplay between dopamine and glutamate in the basal ganglia regulate critical aspects of motor and cognitive behavior. Metabotropic glutamate (mGlu) receptors are key modulators of glutamatergic dysfunction in Parkinson's disease (PD). Preclinical evidence demonstrate that group I mGlu receptor antagonism and groups II and III mGlu receptor activation improve motor symptomatology of PD and decrease l-DOPA-induced dyskinesia by regulating excitatory and inhibitory transmission in the basal ganglia. Emotional and cognitive deficits are also observed in PD. Treatment of these symptoms is challenging and underscore the need for novel effective and well tolerated pharmacological treatments. This article will thus review the currently available knowledge regarding the therapeutic potential of targeting mGlu receptors to restore motor and nonmotor symptoms of PD.
Collapse
Affiliation(s)
- Marianne Amalric
- Aix-Marseille University, CNRS UMR 7291, Laboratoire de Neurosciences Cognitives (LNC), FR3C 3512, 13331 Marseille, France.
| |
Collapse
|
22
|
Rovira X, Harrak Y, Trapero A, González-Bulnes P, Malhaire F, Pin JP, Goudet C, Giraldo J, Llebaria A. Exploring the active conformation of cyclohexane carboxylate positive allosteric modulators of the type 4 metabotropic glutamate receptor. ChemMedChem 2014; 9:2685-98. [PMID: 25196639 DOI: 10.1002/cmdc.201402190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Indexed: 11/07/2022]
Abstract
The active conformation of a family of metabotropic glutamate receptor subtype 4 (mGlu4 ) positive allosteric modulators (PAMs) with the cyclohexane 1,2-dicarboxylic scaffold present in cis-2-(3,5-dichlorophenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041) was investigated by testing structurally similar six-membered ring compounds that have a locked conformation. The norbornane and cyclohexane molecules designed as mGlu4 conformational probes and the enantiomers of the trans diastereomer were computationally characterized and tested in mGlu4 pharmacological assays. The results support a VU0155041 active conformation, with the chair cyclohexane having the aromatic amide substituent in an axial position and the carboxylate in an equatorial position. Moreover, the receptor displays enantiomeric discrimination of the chiral PAMs. The constructed pharmacophore characterized a highly constrained mGlu4 allosteric binding site, thus providing a step forward in structure-based drug design for mGlu4 PAMs.
Collapse
Affiliation(s)
- Xavier Rovira
- Laboratory of Molecular Neuropharmacology & Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193 Bellaterra (Spain); Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier; U661, INSERM, 141 Rue de la Cardonille, 34094 Montpellier (France)
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kalinichev M, Le Poul E, Boléa C, Girard F, Campo B, Fonsi M, Royer-Urios I, Browne SE, Uslaner JM, Davis MJ, Raber J, Duvoisin R, Bate ST, Reynolds IJ, Poli S, Celanire S. Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. J Pharmacol Exp Ther 2014; 350:495-505. [PMID: 24947466 PMCID: PMC4152882 DOI: 10.1124/jpet.114.214437] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/19/2014] [Indexed: 11/22/2022] Open
Abstract
There is growing evidence that activation of metabotropic glutamate receptor 4 (mGlu4) leads to anxiolytic- and antipsychotic-like efficacy in rodent models, yet its relevance to depression-like reactivity remains unclear. Here, we present the pharmacological evaluation of ADX88178 [5-methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine], a novel potent, selective, and brain-penetrant positive allosteric modulator of the mGlu4 receptor in rodent models of anxiety, obsessive compulsive disorder (OCD), fear, depression, and psychosis. ADX88178 dose-dependently reduced the number of buried marbles in the marble burying test and increased open-arm exploration in the elevated plus maze (EPM) test, indicative of anxiolytic-like efficacy. Target specificity of the effect in the EPM test was confirmed using male and female mGlu4 receptor knockout mice. In mice, ADX88178 reduced the likelihood of conditioned freezing in the acquisition phase of the fear conditioning test, yet had no carryover effect in the expression phase. Also, ADX88178 dose-dependently reduced duration of immobility in the forced swim test, indicative of antidepressant-like efficacy. ADX88178 reduced DOI (2,5-dimethoxy-4-iodoamphetamine)-mediated head twitches (albeit with no dose-dependency), and MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine]-induced locomotor hyperactivity in mice, but was inactive in the conditioned avoidance response test in rats. The compound showed good specificity as it had no effect on locomotor activity in mice and rats at efficacious doses. Thus, allosteric activation of mGlu4 receptors can be a promising new therapeutic approach for treatment of anxiety, OCD, fear-related disorders, and psychosis.
Collapse
Affiliation(s)
- Mikhail Kalinichev
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Emmanuel Le Poul
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Christelle Boléa
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Françoise Girard
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Brice Campo
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Massimiliano Fonsi
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Isabelle Royer-Urios
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Susan E Browne
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Jason M Uslaner
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Matthew J Davis
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Jacob Raber
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Robert Duvoisin
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Simon T Bate
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Ian J Reynolds
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Sonia Poli
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Sylvain Celanire
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| |
Collapse
|
24
|
Group III metabotropic glutamate receptors and drug addiction. Front Med 2014; 7:445-51. [PMID: 24078068 DOI: 10.1007/s11684-013-0291-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/05/2013] [Indexed: 02/06/2023]
Abstract
Neuroadaptations of glutamatergic transmission in the limbic reward circuitry are linked to persistent drug addiction. Accumulating data have demonstrated roles of ionotropic glutamate receptors and group I and II metabotropic glutamate receptors (mGluRs) in this event. Emerging evidence also identifies Gαi/o-coupled group III mGluRs (mGluR4/7/8 subtypes enriched in the limbic system) as direct substrates of drugs of abuse and active regulators of drug action. Auto- and heteroreceptors of mGluR4/7/8 reside predominantly on nerve terminals of glutamatergic corticostriatal and GABAergic striatopallidal pathways, respectively. These presynaptic receptors regulate basal and/or phasic release of respective transmitters to maintain basal ganglia homeostasis. In response to operant administration of common addictive drugs, such as psychostimulants (cocaine and amphetamine), alcohol and opiates, limbic group III mGluRs undergo drastic adaptations to contribute to the enduring remodeling of excitatory synapses and to usually suppress drug seeking behavior. As a result, a loss-of-function mutation (knockout) of individual group III receptor subtypes often promotes drug seeking. This review summarizes the data from recent studies on three group III receptor subtypes (mGluR4/7/8) expressed in the basal ganglia and analyzes their roles in the regulation of dopamine and glutamate signaling in the striatum and their participation in the addictive properties of three major classes of drugs (psychostimulants, alcohol, and opiates).
Collapse
|
25
|
Pomierny-Chamioło L, Rup K, Pomierny B, Niedzielska E, Kalivas PW, Filip M. Metabotropic glutamatergic receptors and their ligands in drug addiction. Pharmacol Ther 2014; 142:281-305. [DOI: 10.1016/j.pharmthera.2013.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023]
|
26
|
Gubellini P, Melon C, Dale E, Doller D, Kerkerian-Le Goff L. Distinct effects of mGlu4 receptor positive allosteric modulators at corticostriatal vs. striatopallidal synapses may differentially contribute to their antiparkinsonian action. Neuropharmacology 2014; 85:166-77. [PMID: 24866785 DOI: 10.1016/j.neuropharm.2014.05.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/21/2014] [Accepted: 05/15/2014] [Indexed: 12/20/2022]
Abstract
Metabotropic glutamate 4 (mGlu4) receptor is a promising target for the treatment of motor deficits in Parkinson's disease (PD). This is due in part to its localization at key basal ganglia (BG) synapses that become hyperactive in this pathology, particularly striatopallidal synapses. In this context, mGlu4 receptor activation using either orthosteric agonists or positive allosteric modulators (PAMs) improves motor symptoms in rodent PD models in certain conditions. However, literature data show that mGlu4 receptor PAMs have no effect at striatopallidal GABAergic synapses (unless combined with an orthosteric agonist) and on the firing activity of pallidal neurons, and fail to provide significant motor improvement in relevant PD models. This questions the mechanistic hypothesis that mGlu4 receptor PAMs should act at striatopallidal synapses to alleviate PD motor symptoms. To shed light on this issue, we performed brain slice electrophysiology experiments. We show that Lu AF21934, an mGlu4 PAM small-molecule probe-compound, was ineffective at striatopallidal synapses at all concentrations tested, while it significantly inhibited corticostriatal synaptic transmission. Similarly, Lu AF21934 did not affect electrophysiology readouts at striatopallidal synapses in the presence of haloperidol or in 6-hydroxydopamine-lesioned rats. Interestingly, co-application of Lu AF21934 with a glutamate transporter inhibitor revealed a significant inhibitory action at striatopallidal synapses. Possibly, this effect could rely on increased level/permanence of glutamate in the synaptic cleft. Such differential efficacy of mGlu4 receptor PAMs at corticostriatal vs. striatopallidal synapses raises several issues regarding the synaptic target(s) of these drugs in the BG, and challenges the mechanisms by which they alleviate motor deficits in experimental PD models.
Collapse
Affiliation(s)
- Paolo Gubellini
- Aix-Marseille Université, CNRS, IBDM UMR7288, 13009 Marseille, France.
| | - Christophe Melon
- Aix-Marseille Université, CNRS, IBDM UMR7288, 13009 Marseille, France
| | - Elena Dale
- Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | - Dario Doller
- Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | | |
Collapse
|
27
|
Ossowska K, Wardas J, Berghauzen-Maciejewska K, Głowacka U, Kuter K, Pilc A, Zorn SH, Doller D. Lu AF21934, a positive allosteric modulator of mGlu4 receptors, reduces the harmaline-induced hyperactivity but not tremor in rats. Neuropharmacology 2014; 83:28-35. [PMID: 24726309 DOI: 10.1016/j.neuropharm.2014.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/18/2014] [Accepted: 03/31/2014] [Indexed: 12/19/2022]
Abstract
Harmaline induces tremor in animals resembling essential tremor which has been suggested to result from activation of the glutamatergic olivo-cerebellar projection. The aim of the present study was to examine the effects of systemic administration of Lu AF21934, a brain-penetrating positive allosteric modulator of the metabotropic glutamate receptor 4 (mGlu4), on the harmaline-induced tremor and other forms of motor activity in rats using fully automated Force Plate Actimeters. The influence of harmaline on the mGlu4 mRNA expression in the cerebellum and inferior olive was analysed by in situ hybridization. Harmaline at a dose of 15 mg/kg (ip) triggered tremor which was manifested by an increase in the power within 9-15 Hz band and in the tremor index (a difference in power between bands 9-15 Hz and 0-8 Hz). Harmaline induced a biphasic effect on mobility, initially inhibiting the exploratory locomotor activity of rats (0-30 min after administration), followed by an increase in their basic activity. Lu AF21934 (0.5-5 mg/kg sc) did not influence tremor but at doses of 0.5 and 2.5 mg/kg reversed harmaline-induced hyperactivity. MGlu4 mRNA expression was high in the cerebellar cortex and low in the inferior olive. Repeated harmaline (15 mg/kg ip once a day for 5 days] decreased mGlu4 mRNA in the cerebellum and inferior olive. The present study indicates that the mGlu4 stimulation counteracts hyperactivity induced by harmaline which suggests the involvement of cerebellar glutamatergic transmission in this process. In contrast, neuronal mechanisms involved in tremor seem to be insensitive to the stimulation of mGlu4.
Collapse
Affiliation(s)
- Krystyna Ossowska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland.
| | - Jadwiga Wardas
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Klemencja Berghauzen-Maciejewska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Urszula Głowacka
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Katarzyna Kuter
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 12, Smętna St., 31-343 Kraków, Poland
| | - Stevin H Zorn
- Discovery Chemistry & DMPK, Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | - Dario Doller
- Discovery Chemistry & DMPK, Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| |
Collapse
|
28
|
Jantas D, Greda A, Golda S, Korostynski M, Grygier B, Roman A, Pilc A, Lason W. Neuroprotective effects of metabotropic glutamate receptor group II and III activators against MPP(+)-induced cell death in human neuroblastoma SH-SY5Y cells: the impact of cell differentiation state. Neuropharmacology 2014; 83:36-53. [PMID: 24713472 DOI: 10.1016/j.neuropharm.2014.03.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/25/2014] [Accepted: 03/31/2014] [Indexed: 12/31/2022]
Abstract
Recent studies have documented that metabotropic glutamate receptors from group II and III (mGluR II/III) are a potential target in the symptomatic treatment of Parkinson's disease (PD), however, the neuroprotective effects of particular mGluR II/III subtypes in relation to PD pathology are recognized only partially. In the present study, we investigated the effect of various mGluR II/III activators in the in vitro model of PD using human neuroblastoma SH-SY5Y cell line and mitochondrial neurotoxin MPP(+). We demonstrated that all tested mGluR ligands: mGluR II agonist - LY354740, mGluR III agonist - ACPT-I, mGluR4 PAM - VU0361737, mGluR8 agonist - (S)-3,4-DCPG, mGluR8 PAM - AZ12216052 and mGluR7 allosteric agonist - AMN082 were protective against MPP(+)-evoked cell damage in undifferentiated (UN-) SH-SY5Y cells with the highest neuroprotection mediated by mGluR8-specific agents. However, in retinoic acid- differentiated (RA-) SH-SY5Y cells we found protection mediated only by mGluR8 activators. We also demonstrated the cell proliferation stimulating effect for mGluR4 and mGluR8 PAMs. Next, we showed that the protection mediated by mGluR II/III activators in UN-SH-SY5Y was not accompanied by the modulation of caspase-3 activity, however, a decrease in the number of apoptotic nuclei was found. Finally, we showed that the inhibitor of necroptosis, necrostatin-1 blocked the mGluR III-mediated protection. Altogether our comparative in vitro data add a further proof to neuroprotective effects of mGluR agonists or PAMs and point to mGluR8 as a promising target for neuroprotective interventions in PD. The results also suggest the participation of necroptosis-related molecular pathways in neuroprotective effects of mGluR III activation.
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland.
| | - A Greda
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - S Golda
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - M Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - B Grygier
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - A Roman
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - A Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| | - W Lason
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland
| |
Collapse
|
29
|
Finlay C, Duty S. Therapeutic potential of targeting glutamate receptors in Parkinson's disease. J Neural Transm (Vienna) 2014; 121:861-80. [PMID: 24557498 DOI: 10.1007/s00702-014-1176-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/05/2014] [Indexed: 12/28/2022]
Abstract
Glutamate plays a complex role in many aspects of Parkinson's disease including the loss of dopaminergic neurons, the classical motor symptoms as well as associated non-motor symptoms and the treatment-related side effect, L-DOPA-induced dyskinesia. This widespread involvement opens up possibilities for glutamate-based therapies to provide a more rounded approach to treatment than is afforded by current dopamine replacement therapies. Beneficial effects of blocking postsynaptic glutamate transmission have already been noted in a range of preclinical studies using antagonists of NMDA receptors or negative allosteric modulators of metabotropic glutamate receptor 5 (mGlu5), while positive allosteric modulators of mGlu4 in particular, although at an earlier stage of investigation, also look promising. This review addresses each of the key features of Parkinson's disease in turn, summarising the contribution glutamate makes to that feature and presenting an up-to-date account of the potential for drugs acting at ionotropic or metabotropic glutamate receptors to provide relief. Whilst only a handful of these have progressed to clinical trials to date, notably NMDA and NR2B antagonists against motor symptoms and L-DOPA-induced dyskinesia, with mGlu5 negative allosteric modulators also against L-DOPA-induced dyskinesia, the mainly positive outcomes of these trials, coupled with supportive preclinical data for other strategies in animal models of Parkinson's disease and L-DOPA-induced dyskinesia, raise cautious optimism that a glutamate-based therapeutic approach will have significant impact on the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Clare Finlay
- Wolfson Centre for Age-Related Diseases, King's College London, WW1.28. Hodgkin Building, Guy's Campus, London, SE1 1UL, UK
| | | |
Collapse
|
30
|
Sławińska A, Wierońska JM, Stachowicz K, Marciniak M, Lasoń-Tyburkiewicz M, Gruca P, Papp M, Kusek M, Tokarski K, Doller D, Pilc A. The antipsychotic-like effects of positive allosteric modulators of metabotropic glutamate mGlu4 receptors in rodents. Br J Pharmacol 2014; 169:1824-39. [PMID: 23714045 DOI: 10.1111/bph.12254] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/24/2013] [Accepted: 05/12/2013] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Because agonists at metabotropic glutamate receptors exert beneficial effects in schizophrenia, we have assessed the actions of Lu AF21934 and Lu AF32615, two chemically distinct, selective and brain-penetrant positive allosteric modulators (PAMs) of the mGlu4 receptor, in several tests reflecting positive, negative and cognitive symptoms of schizophrenia in rodents. EXPERIMENTAL APPROACH Hyperactivity induced by MK-801 or amphetamine and head twitches induced by 2,5-dimethoxy-4-iodoamphetamine (DOI) in mice were used as models for positive symptoms. Disruption of social interaction and spatial delayed alternation tests induced by MK-801 in rats were used as models for negative and cognitive symptoms of schizophrenia, respectively. KEY RESULTS Lu AF21934 (0.1-5 mg·kg(-1) ) and Lu AF32615 (2-10 mg·kg(-1) ) dose-dependently inhibited hyperactivity induced by MK-801 or amphetamine. They also antagonized head twitches and increased frequency of spontaneous excitatory postsynaptic currents (EPSCs) in brain slices, induced by DOI. In mice lacking the mGlu4 receptor (mGlu4 (-/-) ) mice, Lu AF21934 did not antagonize DOI-induced head twitches. MK-801-induced disruption in the social interaction test was decreased by Lu AF21934 at 0.5 mg·kg(-1) and by Lu AF32615 at 10 mg·kg(-1) . In the delayed spatial alternation test, Lu AF21934 was active at 1 and 2 mg·kg(-1) , while Lu AF32615 was active at 10 mg·kg(-1) . CONCLUSIONS AND IMPLICATIONS We propose that activation by PAMs of the mGlu4 receptor is a promising approach to the discovery of novel antipsychotic drugs.
Collapse
Affiliation(s)
- Anna Sławińska
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ambrosi G, Cerri S, Blandini F. A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. J Neural Transm (Vienna) 2014; 121:849-59. [DOI: 10.1007/s00702-013-1149-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
|
32
|
Tomiyama K, Kato R, Hara Y, Kobayashi M, Mishina M, Yanagawa Y, Kinsella A, Koshikawa N, Waddington J. Phenotypic characterization of orofacial movement topography in mutants with disruption of amino acid mechanisms: Glutamate N2A/B/D [GluRε1/2/4] subtypes and the GABA synthesizing enzyme GAD65. Neuroscience 2013; 250:743-54. [DOI: 10.1016/j.neuroscience.2013.07.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/02/2013] [Accepted: 07/16/2013] [Indexed: 01/29/2023]
|
33
|
Effects of group II and III metabotropic glutamate receptor ligands on conditioned taste aversion learning. Behav Brain Res 2013; 253:9-16. [DOI: 10.1016/j.bbr.2013.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 06/22/2013] [Accepted: 06/27/2013] [Indexed: 12/13/2022]
|
34
|
Fukuyama K, Tanahashi S, Hoshikawa M, Shinagawa R, Okada M. Zonisamide regulates basal ganglia transmission via astroglial kynurenine pathway. Neuropharmacology 2013; 76 Pt A:137-45. [PMID: 23973311 DOI: 10.1016/j.neuropharm.2013.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 08/01/2013] [Accepted: 08/08/2013] [Indexed: 12/19/2022]
Abstract
To clarify the anti-parkinsonian mechanisms of action of zonisamide (ZNS), we determined the effects of ZNS on tripartite synaptic transmission associated with kynurenine (KYN) pathway (KP) in cultured astrocytes, and transmission in both direct and indirect pathways of basal ganglia using microdialysis. Interactions between cytokines [interferon-γ (IFNγ) and tumor-necrosis factor-α (TNFα)] and ZNS on astroglial releases of KP metabolites, KYN, kynurenic-acid (KYNA), xanthurenic-acid (XTRA), cinnabarinic-acid (CNBA) and quinolinic-acid (QUNA), were determined by extreme liquid-chromatography with mass-spectrometry. Interaction among metabotropic glutamate-receptor (mGluR), KP metabolites and ZNS on striato-nigral, striato-pallidal GABAergic and subthalamo-nigral glutamatergic transmission was examined by microdialysis with extreme liquid-chromatography fluorescence resonance-energy transfer detection. Acute and chronic ZNS administration increased astroglial release of KYN, KYNA, XTRA and CNBA, but not QUNA. Chronic IFNγ administration increased the release of KYN, KYNA, CNBA and QUNA, but had minimal inhibitory effect on XTRA release. Chronic TNFα administration increased CNBA and QUNA, but not KYN, KYNA or XTRA. ZNS inhibited IFNγ-induced elevation of KYN, KYNA and QUNA, but enhanced IFNγ-induced that of CNBA. TNFα-induced rises in CNBA and QUNA were inhibited by ZNS. ZNS inhibited striato-nigral GABAergic, striato-pallidal GABAergic and subthalamo-nigral glutamatergic transmission via activation of groups II and III mGluRs. ZNS enhanced astroglial release of endogenous agonists of group II mGluR, XTRA and group III mGluR, CNBA. Activated endogenous mGluR agonists inhibited transmission in direct and indirect pathways of basal ganglia. These mechanisms contribute to effectiveness and well tolerability of ZNS as an adjunct treatment for Parkinson's disease during l-DOPA monotherapy. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Kouji Fukuyama
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | | | | | | | | |
Collapse
|
35
|
Betts MJ, O'Neill MJ, Duty S. Allosteric modulation of the group III mGlu4 receptor provides functional neuroprotection in the 6-hydroxydopamine rat model of Parkinson's disease. Br J Pharmacol 2012; 166:2317-30. [PMID: 22404342 DOI: 10.1111/j.1476-5381.2012.01943.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE We recently reported that broad spectrum agonist-induced activation of presynaptic group III metabotropic glutamate (mGlu) receptors within the substantia nigra pars compacta using L-2-amino-4-phosphonobutyrate provided functional neuroprotection in the 6-hydroxydopamine lesion rat model of Parkinson's disease. The aim of this study was to establish whether selective activation of the mGlu(4) receptor alone could afford similar functional neuroprotection. EXPERIMENTAL APPROACH The neuroprotective effects of 8 days of supranigral treatment with a positive allosteric modulator of mGlu(4) receptors, (+/-)-cis-2-(3,5-dichlorphenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041), were investigated in rats with unilateral 6-hydroxydopamine lesions. The effects of VU0155041 treatment on motor function were assessed using both habitual (cylinder test) and forced (adjusted stepping, amphetamine-induced rotations) behavioural tests. Nigrostriatal tract integrity was examined by analysis of tyrosine hydroxylase, dopa decarboxylase or dopamine levels in the striatum and tyrosine hydroxylase-positive cell counts in the substantia nigra pars compacta. KEY RESULTS VU0155041 provided around 40% histological protection against a unilateral 6-hydroxydopamine lesion as well as significant preservation of motor function. These effects were inhibited by pre-treatment with (RS)-α-cyclopropyl-4-phosphonophenylglycine, confirming a receptor-mediated response. Reduced levels of inflammatory markers were also evident in the brains of VU0155041-treated animals. CONCLUSIONS AND IMPLICATIONS Allosteric potentiation of mGlu(4) receptors in the substantia nigra pars compacta provided neuroprotective effects in the 6-hydroxydopamine rat model A reduced inflammatory response may contribute, in part, to this action. In addition to the reported symptomatic effects, activation of mGlu(4) receptors may also offer a novel approach for slowing the progressive degeneration observed in Parkinson's disease.
Collapse
Affiliation(s)
- Matthew J Betts
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, UK
| | | | | |
Collapse
|
36
|
Targeting glutamate receptors to tackle the pathogenesis, clinical symptoms and levodopa-induced dyskinesia associated with Parkinson's disease. CNS Drugs 2012; 26:1017-32. [PMID: 23114872 DOI: 10.1007/s40263-012-0016-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The appearance of levodopa-induced dyskinesia (LID) and ongoing degeneration of nigrostriatal dopaminergic neurons are two key features of Parkinson's disease (PD) that current treatments fail to address. Increased glutamate transmission contributes to the motor symptoms in PD, to the striatal plasticity that underpins LID and to the progression of neurodegeneration through excitotoxic mechanisms. Glutamate receptors have therefore long been considered as potential targets for pharmacological intervention in PD, with emphasis on either blocking activation of 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid (AMPA), N-methyl-D-aspartate (NMDA) or excitatory metabotropic glutamate (mGlu) 5 receptors or promoting the activation of group II/III mGlu receptors. Following a brief summary of the role of glutamate in PD and LID, this article explores the current status of pharmacological studies in pre-clinical rodent and primate models through to clinical trials, where applicable, that support the potential of glutamate-based therapeutic interventions. To date, AMPA antagonists have shown good efficacy against LID in rat and primate models, but the failure of perampanel to lessen LID in clinical trials casts doubt on the translational potential of this approach. In contrast, antagonists selective for NR2B-containing NMDA receptors were effective against LID in animal models and in small-scale clinical trials, though observed adverse cognitive effects need addressing. So far, mGlu5 antagonists or negative allosteric modulators (NAMs) look set to become the first introduced for tackling LID, with AFQ-056 reported to exhibit good efficacy in phase II clinical trials. NR2B antagonists and mGlu5 NAMs may subsequently prove to also be effective disease-modifying agents if their protective effects in rat and primate models of PD, respectively, are replicated in the next stages of investigation. Finally, group III mGlu4 agonists or positive allosteric modulators (PAMs), although in the early pre-clinical stages of investigation, are showing good efficacy against motor symptoms, neurodegeneration and LID. It is anticipated that the recent development of mGlu4 PAMs with improved systemic bioavailability will facilitate progression of these agents into the primate model of PD where their potential can be further explored.
Collapse
|
37
|
Le Poul E, Boléa C, Girard F, Poli S, Charvin D, Campo B, Bortoli J, Bessif A, Luo B, Koser AJ, Hodge LM, Smith KM, DiLella AG, Liverton N, Hess F, Browne SE, Reynolds IJ. A potent and selective metabotropic glutamate receptor 4 positive allosteric modulator improves movement in rodent models of Parkinson's disease. J Pharmacol Exp Ther 2012; 343:167-77. [PMID: 22787118 DOI: 10.1124/jpet.112.196063] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Positive allosteric modulators (PAMs) of metabotropic glutamate receptor 4 (mGluR4) have been proposed as a novel therapeutic approach for the treatment of Parkinson's disease. However, evaluation of this proposal has been limited by the availability of appropriate pharmacological tools to interrogate the target. In this study, we describe the properties of a novel mGluR4 PAM. 5-Methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine (ADX88178) enhances glutamate-mediated activation of human and rat mGluR4 with EC(50) values of 4 and 9 nM, respectively. The compound is highly selective for mGluR4 with minimal activities at other mGluRs. Oral administration of ADX88178 in rats is associated with high bioavailability and results in cerebrospinal fluid exposure of >50-fold the in vitro EC(50) value. ADX88178 reverses haloperidol-induced catalepsy in rats at 3 and 10 mg/kg. It is noteworthy that this compound alone has no impact on forelimb akinesia resulting from a bilateral 6-hydroxydopamine lesion in rats. However, coadministration of a low dose of L-DOPA (6 mg/kg) enabled a robust, dose-dependent reversal of the forelimb akinesia deficit. ADX88178 also increased the effects of quinpirole in lesioned rats and enhanced the effects of L-DOPA in MitoPark mice. It is noteworthy that the enhancement of the actions of L-DOPA was not associated with an exacerbation of L-DOPA-induced dyskinesias in rats. ADX88178 is a novel, potent, and selective mGluR4 PAM that is a valuable tool for exploring the therapeutic potential of mGluR4 modulation. The use of this novel tool molecule supports the proposal that activation of mGluR4 may be therapeutically useful in Parkinson's disease.
Collapse
|
38
|
α-Amino-β-fluorocyclopropanecarboxylic acids as a new tool for drug development: Synthesis of glutamic acid analogs and agonist activity towards metabotropic glutamate receptor 4. Bioorg Med Chem 2012; 20:4716-26. [DOI: 10.1016/j.bmc.2012.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 06/01/2012] [Accepted: 06/02/2012] [Indexed: 12/19/2022]
|
39
|
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: 55] [Impact Index Per Article: 4.2] [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
|
40
|
Broadstock M, Austin PJ, Betts MJ, Duty S. Antiparkinsonian potential of targeting group III metabotropic glutamate receptor subtypes in the rodent substantia nigra pars reticulata. Br J Pharmacol 2012; 165:1034-45. [PMID: 21627638 DOI: 10.1111/j.1476-5381.2011.01515.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Increased firing of the glutamatergic pathway between the subthalamic nucleus and substantia nigra pars reticulata (SNpr) contributes to the abnormal firing of motor circuits and subsequent motor deficits seen in Parkinson's disease. Broad spectrum agonist-induced activation of presynaptic group III metabotropic glutamate (mGlu) receptors within the SNpr reduced glutamate release and reversed akinesia in the reserpine-treated rat model of Parkinson's disease. Here, we have sought to identify which subtypes of group III mGlu receptor in the SNpr were responsible for these beneficial effects. EXPERIMENTAL APPROACH The ability of the mGlu(4) positive allosteric modulator, N-phenyl-7-(hydroxyminocyclopropa[b]chromen-1a-carboxamide) (PHCCC), the mGlu(7) allosteric agonist, N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) and the mGlu(8) -selective agonist (S)-3,4-dicarboxyphenylglycine [(S)-3,4-DCPG] to inhibit KCl-evoked [(3) H]-D-aspartate release was examined in vitro in rat nigral prisms. Reversal of akinesia in reserpine-treated rats was also assessed following intranigral injection of these agents. KEY RESULTS PHCCC and AMN082 inhibited [(3) H]-D-aspartate release by 42% and 53%, respectively when given alongside a sub-threshold concentration of the broad spectrum group III agonist, L-2-amino-4-phosphonobutyrate (L-AP4; 1 µM). In contrast (S)-3,4-DCPG failed to inhibit [(3) H]-D-aspartate release. All three agents also reversed reserpine-induced akinesia although only the effects of PHCCC and AMN082 were inhibited by pre-treatment with the group III antagonist (RS)-α-cyclopropyl-4-phosphonophenylglycine (CPPG). CONCLUSIONS AND IMPLICATIONS These findings reveal that targeting SNpr mGlu(4) or mGlu(7) receptors, but not mGlu(8) receptors, provided relief from akinesia in the reserpine-treated rat model of Parkinson's disease, most likely reflecting inhibition of excess glutamate release in this region.
Collapse
Affiliation(s)
- M Broadstock
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, UK
| | | | | | | |
Collapse
|
41
|
Quik M, Park KM, Hrachova M, Mallela A, Huang LZ, McIntosh JM, Grady SR. Role for α6 nicotinic receptors in l-dopa-induced dyskinesias in parkinsonian mice. Neuropharmacology 2012; 63:450-9. [PMID: 22579614 DOI: 10.1016/j.neuropharm.2012.04.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/13/2012] [Accepted: 04/24/2012] [Indexed: 01/09/2023]
Abstract
L-Dopa-induced dyskinesias are a serious side effect that develops in most Parkinson's disease patients on dopamine replacement therapy. Few treatment options are available to manage dyskinesias; however,recent studies show that nicotine reduces these abnormal involuntary movements (AIMs) in parkinsonian animals by acting at nicotinic acetylcholine receptors (nAChRs). Identification of the nAChR subtypes that mediate this reduction in AIMs is important as it will help in the development of nAChR subtype selective drugs for their treatment. Here we investigate the role of α6β2* nAChRs, a subtype selectively present in the nigrostriatal pathway, using a6 nAChR subunit null mutant (α6⁻/⁻) mice.Wildtype and α6⁻/⁻ mice were lesioned by unilateral injection of 6-hydroxydopamine (3 mg/ml) into the medial forebrain bundle. They were then given L-dopa (3 mg/kg) plus benserazide (15 mg/kg) 2e3 wk later. L-dopa-induced AIMs developed to a similar extent in α6⁻/⁻ and wildtype mice.However, AIMs in α6⁻/⁻ mice declined to ~50% of that in wildtype mice with continued L-dopa treatment. Nicotine treatment also decreased AIMs by ~50% in wildtype mice, although not in α6⁻/⁻ mice. There were no effects on parkinsonism under any experimental condition. To conclude, the similar declines in L-dopa-induced AIMs in nicotine-treated wildtype mice and in α6⁻/⁻ mice treated with and without nicotine indicate an essential role for α6β2* nAChRs in the maintenance of L-dopa-induced AIMs.These findings suggest that α6β2* nAChR drugs have potential for reducing L-dopa-induced dyskinesias in Parkinson's disease.
Collapse
Affiliation(s)
- Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Presynaptic mGlu7 receptors control GABA release in mouse hippocampus. Neuropharmacology 2012; 66:215-24. [PMID: 22564442 DOI: 10.1016/j.neuropharm.2012.04.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 04/05/2012] [Accepted: 04/19/2012] [Indexed: 11/24/2022]
Abstract
The functional role of presynaptic release-regulating metabotropic glutamate type 7 (mGlu7) receptors in hippocampal GABAergic terminals was investigated. Mouse hippocampal synaptosomes were preloaded with [(3)H]D-γ-aminobutyric acid ([(3)H]GABA) and then exposed in superfusion to 12 mM KCl. The K(+)-evoked [(3)H]GABA release was inhibited by the mGlu7 allosteric agonist N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082, 0.001-10 μM), as well as by the group III mGlu receptor agonist l-(+)-2-amino-4-phosphonobutyric acid [(l)-AP4, 0.01-1 mM]. The mGlu8 receptor agonist (S)-3,4-dicarboxyphenylglycine [(S)-3,4-DCPG, 10-100 nM] was ineffective. AMN082 and (l)-AP4-induced effects were recovered by the mGlu7 negative allosteric modulator (NAM) 6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP). AMN082 also inhibited in a MMPIP-sensitive manner the K(+)-evoked release of endogenous GABA. AMN082 and the adenylyl cyclase (AC) inhibitor MDL-12,330A reduced [(3)H]GABA exocytosis in a 8-Br-cAMP-sensitive. AMN082-inhibitory effect was additive to that caused by (-)baclofen, but insensitive to the GABA(B) antagonist 3-[[(3,4-Dichlorophenyl)methyl]amino]propyl] diethoxymethyl) phosphinic acid (CGP52432). Conversely, (-)baclofen-induced inhibition of GABA exocytosis was insensitive to MMPIP. Finally, the forskolin-evoked [(3)H]GABA release was reduced by AMN082 or (-)baclofen but abolished when the two agonists were added concomitantly. Mouse hippocampal synaptosomal plasmamembranes posses mGlu7 receptor proteins; confocal microscopy analysis unveiled that mGlu7 proteins colocalize with syntaxin-1A (Stx-1A), with vesicular GABA transporter (VGAT)-proteins and with GABA(B) receptor subunit proteins. We propose that presynaptic inhibitory mGlu7 heteroreceptors, negatively coupled to AC-dependent intraterminal pathway, exist in mouse hippocampal GABA-containing terminals, where they colocalize, but do not functionally cross-talk, with GABA(B) autoreceptors. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
|
43
|
Carbone M, Duty S, Rattray M. Riluzole neuroprotection in a Parkinson's disease model involves suppression of reactive astrocytosis but not GLT-1 regulation. BMC Neurosci 2012; 13:38. [PMID: 22480308 PMCID: PMC3349538 DOI: 10.1186/1471-2202-13-38] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 04/05/2012] [Indexed: 11/10/2022] Open
Abstract
Background Riluzole is a neuroprotective drug used in the treatment of motor neurone disease. Recent evidence suggests that riluzole can up-regulate the expression and activity of the astrocyte glutamate transporter, GLT-1. Given that regulation of glutamate transport is predicted to be neuroprotective in Parkinson's disease, we tested the effect of riluzole in parkinsonian rats which had received a unilateral 6-hydroxydopamine injection into the median forebrain bundle. Results Rats were treated with intraperitoneal riluzole (4 mg/kg or 8 mg/kg), 1 hour before the lesion then once daily for seven days. Riluzole produced a modest but significant attenuation of dopamine neurone degeneration, assessed by suppression of amphetamine-induced rotations, preservation of tyrosine hydroxylase positive neuronal cell bodies in the substantia nigra pars compacta and attenuation of striatal tyrosine hydroxylase protein loss. Seven days after 6-hydroxydopamine lesion, reactive astrocytosis was observed in the striatum, as determined by increases in expression of glial fibrillary acidic protein, however the glutamate transporter, GLT-1, which is also expressed in astrocytes was not regulated by the lesion. Conclusions The results confirm that riluzole is a neuroprotective agent in a rodent model of parkinson's disease. Riluzole administration did not regulate GLT-1 levels but significantly reduced GFAP levels, in the lesioned striatum. Riluzole suppression of reactive astrocytosis is an intriguing finding which might contribute to the neuroprotective effects of this drug.
Collapse
Affiliation(s)
- Marica Carbone
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | | | | |
Collapse
|
44
|
Bennouar KE, Uberti MA, Melon C, Bacolod MD, Jimenez HN, Cajina M, Kerkerian-Le Goff L, Doller D, Gubellini P. Synergy between L-DOPA and a novel positive allosteric modulator of metabotropic glutamate receptor 4: implications for Parkinson's disease treatment and dyskinesia. Neuropharmacology 2012; 66:158-69. [PMID: 22491024 DOI: 10.1016/j.neuropharm.2012.03.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/27/2012] [Accepted: 03/24/2012] [Indexed: 11/29/2022]
Abstract
Group III metabotropic glutamate (mGlu) receptors are localized in presynaptic terminals within basal ganglia (BG) circuitry that become hyperactive due to dopamine depletion in Parkinson's disease (PD). For this reason, group III mGlu receptors, in particular mGlu4, have been considered as key strategic targets for non-dopaminergic pharmacological treatments aimed at modulating these synapses, without producing the well known side-effects of l-DOPA, in particular the highly disabling l-DOPA-induced dyskinesia (LID). Herein we add physiological and functional support to this hypothesis using Lu AF21934, a novel selective and brain-penetrant mGlu4 receptor positive allosteric modulator (PAM) tool compound. By in vitro electrophysiological recordings we demonstrate that Lu AF21934 inhibits corticostriatal synaptic transmission and enhances the effect of the orthosteric mGlu4 receptor-preferred agonist LSP1-2111. In naïve rats, Lu AF21934 dose-dependently (10 and 30 mg/kg) alleviated haloperidol-induced catalepsy. In hemiparkinsonian rats (unilateral 6-hydroxydopamine lesion of the substantia nigra pars compacta), Lu AF21934 alone did not affect akinesia at the doses tested (10 and 30 mg/kg). However, when Lu AF21934 was combined with sub-threshold doses of l-DOPA (1 and 5 mg/kg), it acted synergistically in alleviating akinesia in a dose-dependent manner and, notably, also reduced the incidence of LID but not its severity. Interestingly, these effects occurred at Lu AF21934 brain free concentrations that showed functional activity in in vitro screens (calcium flux and electrophysiology assays). These results support the potential for antiparkinsonian clinical use of a combined treatment consisting in l-DOPA and a mGlu4 receptor PAM to reduce efficacious l-DOPA doses (generally known as l-DOPA sparing), while maintaining the same benefit on PD motor troubles, and at the same time minimizing the development of LID. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Khaled-Ezaheir Bennouar
- Aix-Marseille Univ, Institut de Biologie du Développement de Marseille-Luminy (IBDML), UMR 7288, 13009 Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Goudet C, Vilar B, Courtiol T, Deltheil T, Bessiron T, Brabet I, Oueslati N, Rigault D, Bertrand H, McLean H, Daniel H, Amalric M, Acher F, Pin J. A novel selective metabotropic glutamate receptor 4 agonist reveals new possibilities for developing subtype selective ligands with therapeutic potential. FASEB J 2012; 26:1682-1693. [DOI: 10.1096/fj.11-195941] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Cyril Goudet
- Institut de Génomique FonctionnelleCentre National Recherche Scientifique (CNRS) UMR5203Université de MontpellierMontpellierFrance
- Institut National de la Santé et de la Recherche MédicaleU661MontpellierFrance
| | - Bruno Vilar
- Institut de Génomique FonctionnelleCentre National Recherche Scientifique (CNRS) UMR5203Université de MontpellierMontpellierFrance
- Institut National de la Santé et de la Recherche MédicaleU661MontpellierFrance
| | - Tiphanie Courtiol
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesCNRS UMR8601Université Paris DescartesSorbonne Paris CitéParisFrance
| | - Thierry Deltheil
- Laboratoire de Neurobiologie de la CognitionCNRS UMR6155Aix‐Marseille UniversitéMarseilleFrance
| | - Thomas Bessiron
- Laboratoire de Pharmacologie et Biochimie de la SynapseCNRS UMR8619Institut de Biochimie et de Biophysique Moleculaire et CellulaireUniversité Paris‐Sud 11OrsayFrance
| | - Isabelle Brabet
- Institut de Génomique FonctionnelleCentre National Recherche Scientifique (CNRS) UMR5203Université de MontpellierMontpellierFrance
- Institut National de la Santé et de la Recherche MédicaleU661MontpellierFrance
| | - Nadia Oueslati
- Institut de Génomique FonctionnelleCentre National Recherche Scientifique (CNRS) UMR5203Université de MontpellierMontpellierFrance
- Institut National de la Santé et de la Recherche MédicaleU661MontpellierFrance
| | - Delphine Rigault
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesCNRS UMR8601Université Paris DescartesSorbonne Paris CitéParisFrance
| | | | - Heather McLean
- Laboratoire de Pharmacologie et Biochimie de la SynapseCNRS UMR8619Institut de Biochimie et de Biophysique Moleculaire et CellulaireUniversité Paris‐Sud 11OrsayFrance
| | - Hervé Daniel
- Laboratoire de Pharmacologie et Biochimie de la SynapseCNRS UMR8619Institut de Biochimie et de Biophysique Moleculaire et CellulaireUniversité Paris‐Sud 11OrsayFrance
| | - Marianne Amalric
- Laboratoire de Neurobiologie de la CognitionCNRS UMR6155Aix‐Marseille UniversitéMarseilleFrance
| | - Francine Acher
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesCNRS UMR8601Université Paris DescartesSorbonne Paris CitéParisFrance
| | - Jean‐Philippe Pin
- Institut de Génomique FonctionnelleCentre National Recherche Scientifique (CNRS) UMR5203Université de MontpellierMontpellierFrance
- Institut National de la Santé et de la Recherche MédicaleU661MontpellierFrance
| |
Collapse
|
46
|
Dickerson JW, Conn PJ. Therapeutic potential of targeting metabotropic glutamate receptors for Parkinson's disease. Neurodegener Dis Manag 2012; 2:221-232. [PMID: 23526920 DOI: 10.2217/nmt.12.6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disorder predominantly characterized by motor symptoms including bradykinesia and resting tremor. The gold standard of treatment for PD remains dopamine replacement therapy, which eventually fails due to continued progression of the disease and the development of debilitating side effects. Recent breakthroughs are providing the first major advances in the development of fundamentally new pharmacological strategies for the treatment of PD that do not rely on dopamine replacement strategies, but rather aim to reduce the overactive indirect pathway within the basal ganglia. In this article, we will review the role of metabotropic glutamate receptors within the basal ganglia and discuss the potential for modulation of metabotropic glutamate receptors as a treatment for PD.
Collapse
Affiliation(s)
- Jonathan W Dickerson
- Vanderbilt University Medical Center, Department of Pharmacology & Center for Neuroscience Drug Discovery, 1205 LH, Nashville, TN 37232, USA
| | | |
Collapse
|
47
|
Kramer T, Lo Monte F, Göring S, Okala Amombo GM, Schmidt B. Small molecule kinase inhibitors for LRRK2 and their application to Parkinson's disease models. ACS Chem Neurosci 2012; 3:151-60. [PMID: 22860184 PMCID: PMC3369800 DOI: 10.1021/cn200117j] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/18/2012] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Several single gene mutations have been linked to this disease. Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) indicate LRRK2 as promising therapeutic target for the treatment of PD. LRRK2 mutations were observed in sporadic as well as familial PD patients and have been investigated intensively. LRRK2 is a large and complex protein, with multiple enzymatic and protein-interaction domains, each of which is effected by mutations. The most common mutation in PD patients is G2019S. Several LRRK2 inhibitors have been reported already, although the crystal structure of LRRK2 has not yet been determined. This review provides a summary of known LRRK2 inhibitors and will discuss recent in vitro and in vivo results of these inhibitors.
Collapse
Affiliation(s)
| | | | - Stefan Göring
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Ghislaine Marlyse Okala Amombo
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Boris Schmidt
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
48
|
Célanire S, Campo B. Recent advances in the drug discovery of metabotropic glutamate receptor 4 (mGluR4) activators for the treatment of CNS and non-CNS disorders. Expert Opin Drug Discov 2012; 7:261-80. [PMID: 22468956 DOI: 10.1517/17460441.2012.660914] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The metabotropic glutamate receptor type 4 (mGluR4) plays a pivotal role in a plethora of therapeutic areas, as recently demonstrated in preclinical validation studies with several chemical classes of compounds in rodent models of central nervous system (CNS) and peripheral disorders. Activation of mGluR4 with orthosteric agonists, allosteric agonists or pure positive allosteric modulators (PAM) has been postulated to be of broad therapeutic use. AREAS COVERED The authors address past and current drug discovery efforts, insights and achievements in the field toward the identification of therapeutically promising and emerging class of mGluR4 activators, over the 2005 - 2011 period. Chemical structures, properties and in vivo pharmacological results discussed in the present review were retrieved from public literature including PubMed searches, Thomson Pharma and SciFinder databases searches, conferences, proceedings and posters. EXPERT OPINION Developing a subtype-selective, orally bioavailable brain penetrant mGluR4 orthosteric agonist remains challenging. Lack of subtype selectivity and low brain penetration has been a common limitation of the first generation of mGluR4 agonist and potentiators. However, significant progress has recently been made with the identification of several double- to single-digit nanomolar mGluR4 PAM having reasonable pharmacokinetic properties, oral bioavailability and brain penetration. The use of such compounds in research has led to advancement in understanding the central role of mGluR4 in multiple neurodegenerative and neuroinflammatory disorders, such as Parkinson's disease and multiple sclerosis. Our understanding of the potential application of mGluR4 as therapeutic target is expected to grow as these compounds advance into preclinical and clinical development.
Collapse
Affiliation(s)
- Sylvain Célanire
- Medicinal Chemistry Department, Addex Pharmaceuticals, Geneva, Switzerland.
| | | |
Collapse
|
49
|
Metabotropic glutamate receptors in neurodegeneration/neuroprotection: still a hot topic? Neurochem Int 2012; 61:559-65. [PMID: 22306345 DOI: 10.1016/j.neuint.2012.01.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 01/14/2012] [Accepted: 01/14/2012] [Indexed: 01/17/2023]
Abstract
Moving from early studies, we here review the most recent evidence linking metabotropic glutamate (mGlu) receptors to processes of neurodegeneration/neuroprotection. The use of knockout mice and subtype-selective drugs has increased our knowledge of the precise role played by individual mGlu receptor subtypes in these processes. Activation of mGlu1 and mGlu5 receptors may either amplify or reduce neuronal damage depending on the context and the nature of the toxic insults. In contrast, mGlu1 and mGlu5 receptors antagonists are consistently protective in in vitro and in vivo models of neuronal death. A series of studies suggest that mGlu1 receptor antagonists or negative allosteric modulators (NAMs) are promising candidates for the treatment of ischemic brain damage, whereas mGlu5 receptor NAMs, which have been clinically developed for the treatment of Parkinson's disease (PD) and l-DOPA-induced dyskinesias, protect nigro-striatal dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity in mice and monkeys. Activation of glial mGlu3 receptors promotes the formation of various neurotrophic factors, such as transforming growth factor-β (TGF-β), glial-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF). Hence, selective mGlu3 receptor agonists or positive allosteric modulators (PAMs) (not yet available) are potentially helpful in the treatment of chronic neurodegenerative disorders such as PD, Alzheimer's disease (AD), and amyotrophic lateral sclerosis. Selective mGlu2 receptor PAMs should be used with caution in AD patients because these drugs are shown to amplify β-amyloid neurotoxicity. Finally, mGlu4 receptor agonists/PAMs share with mGlu5 receptor NAMs the ability to improve motor symptoms associated with PD and attenuate nigro-striatal degeneration at the same time. No data are yet available on the role of mGlu7 and mGlu8 receptors in neurodegeneration/neuroprotection.
Collapse
|
50
|
Lopez S, Jouve L, Turle-Lorenzo N, Kerkerian-Legoff L, Salin P, Amalric M. Antiparkinsonian action of a selective group III mGlu receptor agonist is associated with reversal of subthalamonigral overactivity. Neurobiol Dis 2012; 46:69-77. [PMID: 22245662 DOI: 10.1016/j.nbd.2011.12.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/14/2011] [Accepted: 12/20/2011] [Indexed: 01/02/2023] Open
Abstract
Activation of group III metabotropic glutamate (mGlu) receptors has been recently highlighted as a potential approach in the treatment of Parkinson's disease (PD). This study evaluates the antiparkinsonian action of systemic administration of the broad-spectrum agonist of group III mGlu receptors, 1-aminocyclopentane-1,3,4-tricarboxylic acid (ACPT-I), and investigates its site of action within the basal ganglia circuitry. Acute injection of ACPT-I reverses haloperidol-induced catalepsy, an index of akinesia in rodents. In a rat model of early PD based on partial bilateral nigrostriatal lesions, chronic (2weeks) administration of ACPT-I is required to efficiently alleviate the akinetic deficit evidenced in a reaction time task. This treatment counteracts the post-lesional increases in the gene expression of cytochrome oxidase subunit I, a metabolic marker of neuronal activity, in the overall subthalamic nucleus and in the lateral motor part of the substantia nigra pars reticulata (SNr) but has no effect in the globus pallidus. Paradoxically, ACPT-I administration in sham animals impairs performance and induces overexpression of cytochrome oxidase subunit I mRNA in the lateral SNr, and has no effect in the subthalamic nucleus or globus pallidus. Altogether, our results provide new evidence for the antiparkinsonian efficiency of group III mGlu receptor agonism, point to the regulation of the overactive subthalamo-nigral connection as a main site of action in an early stage of PD and underline the complex interplay between these receptors and the dopaminergic system to regulate basal ganglia function in control and PD conditions.
Collapse
Affiliation(s)
- S Lopez
- Aix-Marseille Univ, Laboratoire de Neurobiologie de la Cognition, 13331 Marseille Cx 3, France
| | | | | | | | | | | |
Collapse
|