1
|
Bourque M, Morissette M, Isenbrandt A, Giatti S, Melcangi RC, Carta M, Frau R, Bortolato M, Soulet D, Di Paolo T. Effect of 5-alpha reductase inhibitors in animal models of Parkinson's disease. Front Neuroendocrinol 2024; 75:101156. [PMID: 39353534 DOI: 10.1016/j.yfrne.2024.101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD) is characterized by motor symptoms due to loss of brain dopamine and non-motor symptoms, including gastrointestinal disorders. Although there is no cure for PD, symptomatic treatments are available. L-Dopa is the gold standard PD therapy, but most patients develop dyskinesias (LID), which are challenging to manage. Amantadine is recognized as the most effective drug for LID, but its adverse effects limit the use in patients. Here we review how 5α-reductase inhibitors (5ARIs), drugs used to treat benign prostatic hyperplasia and alopecia, exhibit beneficial effects in PD animal models. 5ARIs show neuroprotective properties in brain and gut dopaminergic systems, and reduce dyskinesias in rodent model of PD. Additionally, the 5ARI finasteride dampened dopaminergic-induced drug gambling in PD patients. Neuroprotection and antidyskinetic activities of 5ARIs in animal models of PD suggest their potential repurposing in men with PD to address gut dysfunction, protect brain DA and inhibit dyskinesias.
Collapse
Affiliation(s)
- Mélanie Bourque
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Amandine Isenbrandt
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Denis Soulet
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
2
|
Cardinale A, de Iure A, Picconi B. Neuroinflammation and Dyskinesia: A Possible Causative Relationship? Brain Sci 2024; 14:514. [PMID: 38790492 PMCID: PMC11118841 DOI: 10.3390/brainsci14050514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
Levodopa (L-DOPA) treatment represents the gold standard therapy for Parkinson's disease (PD) patients. L-DOPA therapy shows many side effects, among them, L-DOPA-induced dyskinesias (LIDs) remain the most problematic. Several are the mechanisms underlying these processes: abnormal corticostriatal neurotransmission, pre- and post-synaptic neuronal events, changes in gene expression, and altered plasticity. In recent years, researchers have also suggested non-neuronal mechanisms as a possible cause for LIDs. We reviewed recent clinical and pre-clinical studies on neuroinflammation contribution to LIDs. Microglia and astrocytes seem to play a strategic role in LIDs phenomenon. In particular, their inflammatory response affects neuron-glia communication, synaptic activity and neuroplasticity, contributing to LIDs development. Finally, we describe possible new therapeutic interventions for dyskinesia prevention targeting glia cells.
Collapse
Affiliation(s)
- Antonella Cardinale
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Antonio de Iure
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Barbara Picconi
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| |
Collapse
|
3
|
Bourque M, Morissette M, Di Paolo T. Neuroactive steroids and Parkinson's disease: Review of human and animal studies. Neurosci Biobehav Rev 2024; 156:105479. [PMID: 38007170 DOI: 10.1016/j.neubiorev.2023.105479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The greater prevalence and incidence of Parkinson's disease (PD) in men suggest a beneficial effect of sex hormones. Neuroactive steroids have neuroprotective activities thus offering interesting option for disease-modifying therapy for PD. Neuroactive steroids are also neuromodulators of neurotransmitter systems and may thus help to control PD symptoms and side effect of dopamine medication. Here, we review the effect on sex hormones (estrogen, androgen, progesterone and its metabolites) as well as androstenediol, pregnenolone and dehydroepiandrosterone) in human studies and in animal models of PD. The effect of neuroactive steroids is reviewed by considering sex and hormonal status to help identify specifically for women and men with PD what might be a preventive approach or a symptomatic treatment. PD is a complex disease and the pathogenesis likely involves multiple cellular processes. Thus it might be useful to target different cellular mechanisms that contribute to neuronal loss and neuroactive steroids provide therapeutics options as they have multiple mechanisms of action.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada; Faculté de pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec G1V 0A6, Canada.
| |
Collapse
|
4
|
Gautam D, Naik UP, Naik MU, Yadav SK, Chaurasia RN, Dash D. Glutamate Receptor Dysregulation and Platelet Glutamate Dynamics in Alzheimer's and Parkinson's Diseases: Insights into Current Medications. Biomolecules 2023; 13:1609. [PMID: 38002291 PMCID: PMC10669830 DOI: 10.3390/biom13111609] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Two of the most prevalent neurodegenerative disorders (NDDs), Alzheimer's disease (AD) and Parkinson's disease (PD), present significant challenges to healthcare systems worldwide. While the etiologies of AD and PD differ, both diseases share commonalities in synaptic dysfunction, thereby focusing attention on the role of neurotransmitters. The possible functions that platelets may play in neurodegenerative illnesses including PD and AD are becoming more acknowledged. In AD, platelets have been investigated for their ability to generate amyloid-ß (Aß) peptides, contributing to the formation of neurotoxic plaques. Moreover, platelets are considered biomarkers for early AD diagnosis. In PD, platelets have been studied for their involvement in oxidative stress and mitochondrial dysfunction, which are key factors in the disease's pathogenesis. Emerging research shows that platelets, which release glutamate upon activation, also play a role in these disorders. Decreased glutamate uptake in platelets has been observed in Alzheimer's and Parkinson's patients, pointing to a systemic dysfunction in glutamate handling. This paper aims to elucidate the critical role that glutamate receptors play in the pathophysiology of both AD and PD. Utilizing data from clinical trials, animal models, and cellular studies, we reviewed how glutamate receptors dysfunction contributes to neurodegenerative (ND) processes such as excitotoxicity, synaptic loss, and cognitive impairment. The paper also reviews all current medications including glutamate receptor antagonists for AD and PD, highlighting their mode of action and limitations. A deeper understanding of glutamate receptor involvement including its systemic regulation by platelets could open new avenues for more effective treatments, potentially slowing disease progression and improving patient outcomes.
Collapse
Affiliation(s)
- Deepa Gautam
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Ulhas P. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Meghna U. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Santosh K. Yadav
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Rameshwar Nath Chaurasia
- The Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India;
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
5
|
Herrero MT, Yuste JE, Cuenca-Bermejo L, Almela P, Arenas-Betancur L, De Pablos V, Gonzalez-Cuello A, Del Bel E, Navarro-Zaragoza J, Fernández-Villalba E. 7-Nitroindazole reduces L-DOPA-induced dyskinesias in non-human Parkinsonian primate. Open Biol 2023; 13:220370. [PMID: 37192671 DOI: 10.1098/rsob.220370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/31/2023] [Indexed: 05/18/2023] Open
Abstract
Nitric oxide (NO) plays a pivotal role in integrating dopamine transmission in the basal ganglia and has been implicated in the pathogenesis of Parkinson disease (PD). The objective of this study was to ascertain whether the NO synthase inhibitor, 7-nitroindazole (7-NI), is able to reduce L-DOPA-induced dyskinesias (LIDs) in a non-human primate model of PD chronically intoxicated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Six Parkinsonian macaques were treated daily with L-DOPA for 3-4 months until they developed LIDs. Three animals were then co-treated with a single dose of 7-NI administered 45 min before each L-DOPA treatment. Dyskinetic MPTP-treated monkeys showed a significant decrease in LIDs compared with their scores without 7-NI treatment (p < 0.05). The anti-Parkinsonian effect of L-DOPA was similar in all three monkeys with and without 7-NI co-treatment. This improvement was significant with respect to the intensity and duration of LIDs while the beneficial effect of L-DOPA treatment was maintained and could represent a promising therapy to improve the quality of life of PD patients.
Collapse
Affiliation(s)
- M T Herrero
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - J E Yuste
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
| | - L Cuenca-Bermejo
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - P Almela
- Department of Pharmacology, School of Medicine, University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - L Arenas-Betancur
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
| | - V De Pablos
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - A Gonzalez-Cuello
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - E Del Bel
- Department of Basic and Oral Biology, Faculty of Odontology of Ribeirão Preto (FORP-USP) and Center for Research Support on Applied Neuroscience (NAPNA-USP), University of São Paulo, Ribeirão Preto, SP 14040-904, Brazil
| | - J Navarro-Zaragoza
- Department of Pharmacology, School of Medicine, University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| | - E Fernández-Villalba
- Clinical and Experimental Neuroscience (NiCE), Institute for Aging Research, School of Medicine, Campus Mare Nostrum, The European University for Well-Being, EUniWell, University of Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
6
|
Frouni I, Huot P. Glutamate modulation for the treatment of levodopa induced dyskinesia: a brief review of the drugs tested in the clinic. Neurodegener Dis Manag 2022; 12:203-214. [PMID: 35587024 DOI: 10.2217/nmt-2021-0055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Levodopa is the standard treatment for Parkinson's disease, but its use is marred by the emergence of dyskinesia, for which treatment options remain limited. Here, we review the glutamatergic modulators that were assessed for their antidyskinetic potential in clinical trials, including N-methyl-D-aspartate (NMDA) antagonists, agonists at the glycine-binding site on NMDA receptors, metabotropic glutamate (mGlu) 4 agonists, mGlu5 antagonists, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonists and glutamate release inhibitors. Several agents that were investigated are not selective for their targets, raising uncertainty about the extent to which glutamatergic modulation contributed to their effects. Except for amantadine, the use of glutamatergic modulators for the treatment of dyskinesia in Parkinson's disease remains largely investigational, with promising results obtained with mGlu5 negative allosteric modulation.
Collapse
Affiliation(s)
- Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, H3C 3J7, Canada.,Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Neuroscience, Division of Neurology, McGill University Health Centre, Montreal, QC, H3A 2B4, Canada
| |
Collapse
|
7
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Dhuriya YK, Sharma D. Neuronal Plasticity: Neuronal Organization is Associated with Neurological Disorders. J Mol Neurosci 2020; 70:1684-1701. [PMID: 32504405 DOI: 10.1007/s12031-020-01555-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022]
Abstract
Stimuli from stressful events, attention in the classroom, and many other experiences affect the functionality of the brain by changing the structure or reorganizing the connections between neurons and their communication. Modification of the synaptic transmission is a vital mechanism for generating neural activity via internal or external stimuli. Neuronal plasticity is an important driving force in neuroscience research, as it is the basic process underlying learning and memory and is involved in many other functions including brain development and homeostasis, sensorial training, and recovery from brain injury. Indeed, neuronal plasticity has been explored in numerous studies, but it is still not clear how neuronal plasticity affects the physiology and morphology of the brain. Thus, unraveling the molecular mechanisms of neuronal plasticity is essential for understanding the operation of brain functions. In this timeline review, we discuss the molecular mechanisms underlying different forms of synaptic plasticity and their association with neurodegenerative/neurological disorders as a consequence of alterations in neuronal plasticity.
Collapse
Affiliation(s)
- Yogesh Kumar Dhuriya
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR) Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, India
| | - Divakar Sharma
- Department of Biochemistry, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India.
- CRF, Mass Spectrometry Laboratory, Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology-Delhi (IIT-D), Delhi, 110016, India.
| |
Collapse
|
9
|
Olanow CW, Calabresi P, Obeso JA. Continuous Dopaminergic Stimulation as a Treatment for Parkinson's Disease: Current Status and Future Opportunities. Mov Disord 2020; 35:1731-1744. [DOI: 10.1002/mds.28215] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- C. Warren Olanow
- Department of Neurology and Department of Neuroscience Mount Sinai School of Medicine New York New York USA
- Clintrex Research Corporation Sarasota Florida USA
| | - Paolo Calabresi
- Neurology Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
- Dipartimento Neuroscienze Università Cattolica del Sacro Cuore Rome Italy
| | - Jose A. Obeso
- CINAC, Hospital Universitario HM Puerta del Sur, Universidad CEU‐San Pablo Móstoles Madrid Spain
- CIBERNED, Instituto de Salud Carlos III Madrid Spain
| |
Collapse
|
10
|
AlShimemeri S, Fox SH, Visanji NP. Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update. Expert Opin Emerg Drugs 2020; 25:131-144. [DOI: 10.1080/14728214.2020.1763954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sohaila AlShimemeri
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Susan H Fox
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
11
|
Boi L, Pisanu A, Greig NH, Scerba MT, Tweedie D, Mulas G, Fenu S, Carboni E, Spiga S, Carta AR. Immunomodulatory drugs alleviate l-dopa-induced dyskinesia in a rat model of Parkinson's disease. Mov Disord 2019; 34:1818-1830. [PMID: 31335998 PMCID: PMC11719776 DOI: 10.1002/mds.27799] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/05/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Thalidomide and closely related analogues are used clinically for their immunomodulatory and antiangiogenic properties mediated by the inhibition of the proinflammatory cytokine tumor necrosis factor α. Neuroinflammation and angiogenesis contribute to classical neuronal mechanisms underpinning the pathophysiology of l-dopa-induced dyskinesia, a motor complication associated with l-dopa therapy in Parkinson's disease. The efficacy of thalidomide and the more potent derivative 3,6'-dithiothalidomide on dyskinesia was tested in the 6-hydroxydopamine Parkinson's disease model. METHODS Three weeks after 6-hydroxydopamine infusion, rats received 10 days of treatment with l-dopa plus benserazide (6 mg/kg each) and thalidomide (70 mg/kg) or 3,6'-dithiothalidomide (56 mg/kg), and dyskinesia and contralateral turning were recorded daily. Rats were euthanized 1 hour after the last l-dopa injection, and levels of tumor necrosis factor-α, interleukin-10, OX-42, vimentin, and vascular endothelial growth factor immunoreactivity were measured in their striatum and substantia nigra reticulata to evaluate neuroinflammation and angiogenesis. Striatal levels of GLUR1 were measured as a l-dopa-induced postsynaptic change that is under tumor necrosis factor-α control. RESULTS Thalidomide and 3,6'-dithiothalidomide significantly attenuated the severity of l-dopa-induced dyskinesia while not affecting contralateral turning. Moreover, both compounds inhibited the l-dopa-induced microgliosis and excessive tumor necrosis factor-α in the striatum and substantia nigra reticulata, while restoring physiological levels of the anti-inflammatory cytokine interleukin-10. l-Dopa-induced angiogenesis was inhibited in both basal ganglia nuclei, and l-dopa-induced GLUR1 overexpression in the dorsolateral striatum was restored to normal levels. CONCLUSIONS These data suggest that decreasing tumor necrosis factor-α levels may be useful to reduce the appearance of dyskinesia, and thalidomide, and more potent derivatives may provide an effective therapeutic approach to dyskinesia. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Augusta Pisanu
- CNR Institute of Neuroscience, Cagliari, Cagliari, Italy
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Michael T. Scerba
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Sandro Fenu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
12
|
Roles of Glutamate Receptors in Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20184391. [PMID: 31500132 PMCID: PMC6769661 DOI: 10.3390/ijms20184391] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder resulting from the degeneration of pigmented dopaminergic neurons in the substantia nigra pars compacta. It induces a series of functional modifications in the circuitry of the basal ganglia nuclei and leads to severe motor disturbances. The amino acid glutamate, as an excitatory neurotransmitter, plays a key role in the disruption of normal basal ganglia function regulated through the interaction with its receptor proteins. It has been proven that glutamate receptors participate in the modulation of neuronal excitability, transmitter release, and long-term synaptic plasticity, in addition to being related to the altered neurotransmission in Parkinson's disease. Therefore, they are considered new targets for improving the therapeutic strategies used to treat Parkinson's disease. In this review, we discuss the biological characteristics of these receptors and demonstrate the receptor-mediated neuroprotection in Parkinson's disease. Pharmacological manipulation of these receptors during anti-Parkinsonian processes in both experimental studies and clinical trials are also summarized.
Collapse
|
13
|
Chang Y, Du C, Han L, Lv S, Zhang J, Bian G, Tang G, Liu Y, Chen T, Liu J. Enhanced AMPA receptor-mediated excitatory transmission in the rodent rostromedial tegmental nucleus following lesion of the nigrostriatal pathway. Neurochem Int 2018; 122:85-93. [PMID: 30448565 DOI: 10.1016/j.neuint.2018.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/23/2018] [Accepted: 11/09/2018] [Indexed: 11/24/2022]
Abstract
The GABAergic rostromedial tegmental nucleus (RMTg) has reciprocal connections with the dopaminergic ventral tegmental area and substantia nigra pars compacta (SNc), and is involved in inhibitory control of monoaminergic nuclei. At present, it is not clear whether unilateral 6-hydroxydopamine lesions of the SNc in rats affect AMPA receptor-mediated excitatory transmission in the RMTg. Here we found that lesions of the SNc in rats increased the firing rate of GABAergic neurons and the level of glutamate in the RMTg compared to sham-operated rats. Intra-RMTg injection of AMPA receptor agonist (S)-AMPA increased the firing rate of the GABAergic neurons in both sham-operated and the lesioned rats, while AMPA receptor antagonist NBQX decreased the firing rate of the neurons. Further, intra-RMTg injection of (S)-AMPA decreased the levels of dopamine and serotonin in the medial prefrontal cortex (mPFC) in the two groups of rats; conversely, NBQX increased the levels of dopamine and serotonin. Compared to sham-operated rats, the duration of (S)-AMPA and NBQX action on the firing rate of GABAergic neurons in the RMTg and release of doapmine and serotonin in the mPFC was prolonged in the lesioned rats. In addition, lesions of the SNc in rats increased protein expression of t-GluR1 and p-GluR1-S831 subunits compared to sham-operated rats. Therefore, these changes in the lesioned rats are associated with increased release of glutamate and up-regulated expression of GluR1 subunit-containing AMPA receptors in the RMTg, which suggest that degeneration of the nigrostriatal pathway enhances AMPA receptor-mediated excitatory transmission in the RMTg.
Collapse
Affiliation(s)
- Yongli Chang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Physiology, Changzhi Medical College, Changzhi, 046000, China
| | - Chengxue Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Lingna Han
- Department of Physiology, Changzhi Medical College, Changzhi, 046000, China
| | - Shuxuan Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Guanyun Bian
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Guoyi Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yiwei Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Tao Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
14
|
Dragašević-Mišković N, Petrović I, Stanković I, Kostić VS. Chemical management of levodopa-induced dyskinesia in Parkinson's disease patients. Expert Opin Pharmacother 2018; 20:219-230. [PMID: 30411647 DOI: 10.1080/14656566.2018.1543407] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Levodopa-induced dyskinesias (LID) appears in more than 50% of Parkinson's disease patients after 5 years of treatment and clinicians always have to ensure that there is a balance between the beneficial effect of the treatment and the potential complications. AREAS COVERED In this review, the authors discuss the treatment of LID. Treatment can be divided into strategies for preventing their occurrence, modification of dopaminergic therapy, and providing more continuous dopaminergic stimulation as well as the use of nondopaminergic drugs. EXPERT OPINION Amantadine is currently considered the most effective drug for the treatment of LID. Several compounds developed to target adenosine, adrenergic, glutamatergic, and serotonergic receptors have shown to significantly decrease dyskinesias in animal models. However, despite promising preclinical results, translation to clinical practice remains challenging and majority of these compounds failed to decrease LID in randomized controlled trials with moderate-to-advanced parkinsonian patients. Despite promising results with nondopaminergic drugs, treatment of dyskinesias is still challenging and largely due to their side effects. Future research should focus on developing treatments that can provide continuous dopaminergic delivery throughout the day in a noninvasive manner. Studies on the impact of the early administration of long-acting formulations of levo-3,4-dihydroxy-phenylalanine on dyskinesias are also necessary.
Collapse
Affiliation(s)
| | - Igor Petrović
- a Neurology Clinic, CCS, School of Medicine , Universtiy of Belgrade , Belgrade , Serbia
| | - Iva Stanković
- a Neurology Clinic, CCS, School of Medicine , Universtiy of Belgrade , Belgrade , Serbia
| | - Vladimir S Kostić
- a Neurology Clinic, CCS, School of Medicine , Universtiy of Belgrade , Belgrade , Serbia
| |
Collapse
|
15
|
Veyres N, Hamadjida A, Huot P. Predictive Value of Parkinsonian Primates in Pharmacologic Studies: A Comparison between the Macaque, Marmoset, and Squirrel Monkey. J Pharmacol Exp Ther 2018; 365:379-397. [PMID: 29523699 DOI: 10.1124/jpet.117.247171] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/06/2018] [Indexed: 03/08/2025] Open
Abstract
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate is the gold-standard animal model of Parkinson disease (PD) and has been used to assess the effectiveness of experimental drugs on dyskinesia, parkinsonism, and psychosis. Three species have been used in most studies-the macaque, marmoset, and squirrel monkey-the last much less so than the first two species; however, the predictive value of each species at forecasting clinical efficacy, or lack thereof, is poorly documented. Here, we have reviewed all the published literature detailing pharmacologic studies that assessed the effects of experimental drugs on dyskinesia, parkinsonism, and psychosis in each of these species and have calculated their predictive value of success and failure at the clinical level. We found that, for dyskinesia, the macaque has a positive predictive value of 87.5% and a false-positive rate of 38.1%, whereas the marmoset has a positive predictive value of 76.9% and a false-positive rate of 15.6%. For parkinsonism, the macaque has a positive predictive value of 68.2% and a false-positive rate of 44.4%, whereas the marmoset has a positive predictive value of 86.9% and a false-positive rate of 41.7%. No drug that alleviates psychosis in the clinic has shown efficacy at doing so in the macaque, whereas the marmoset has 100% positive predictive value. The small number of studies conducted in the squirrel monkey precluded us from calculating its predictive efficacy. We hope our results will help in the design of pharmacologic experiments and will facilitate the drug discovery and development process in PD.
Collapse
Affiliation(s)
- Nicolas Veyres
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| | - Adjia Hamadjida
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| | - Philippe Huot
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (N.V.),Montreal Neurological Institute (A.H.,P.H.), and Department of Neurology and Neurosurgery, McGill University (P.H.), Montreal, Quebec, Canada
| |
Collapse
|
16
|
Cerri S, Siani F, Blandini F. Investigational drugs in Phase I and Phase II for Levodopa-induced dyskinesias. Expert Opin Investig Drugs 2017; 26:777-791. [PMID: 28535734 DOI: 10.1080/13543784.2017.1333598] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Prolonged treatment of Parkinson's disease (PD) with levodopa (L-DOPA) results in motor complications, including motor fluctuations and involuntary movements known as L-DOPA induced dyskinesias (LIDs). LIDs represent an additional cause of disability for PD patients and a major challenge for the clinical neurologist. Preclinical research has provided invaluable insights into the molecular and neural substrates of LIDs, identifying a number of potential targets for new anti-dyskinetic strategies. Areas covered: This review article is centered on drugs currently in Phase I and II clinical trials for LIDs and their relative pharmacological targets, which include glutamate, acetylcholine, serotonin, adrenergic receptors and additional targets of potential therapeutic interest. Expert opinion: LIDs are sustained by complex molecular and neurobiological mechanisms that are difficult to disentangle or target, unless one or more prevalent mechanisms are identified. In this context, the role of the serotonergic system and mGluR5 glutamate receptors seem to stand out. Interesting results have been obtained, for example, with partial 5-HT1A/5-HT1B receptor agonist eltoprazine and mGluR5 negative allosteric modulator dipraglurant. Confirmation of these results through large-scale, Phase III clinical trials will be needed, to obtain new pharmacological tools that may be used to optimize the treatment of PD patients with motor complications.
Collapse
Affiliation(s)
- Silvia Cerri
- a Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases , C. Mondino National Neurological Institute , Pavia , Italy
| | - Francesca Siani
- a Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases , C. Mondino National Neurological Institute , Pavia , Italy
| | - Fabio Blandini
- a Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases , C. Mondino National Neurological Institute , Pavia , Italy
| |
Collapse
|
17
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|
18
|
Morin N, Morissette M, Grégoire L, Rajput A, Rajput AH, Di Paolo T. Contribution of brain serotonin subtype 1B receptors in levodopa-induced motor complications. Neuropharmacology 2015; 99:356-68. [PMID: 26254863 DOI: 10.1016/j.neuropharm.2015.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/03/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022]
Abstract
L-DOPA-induced dyskinesias (LID) are abnormal involuntary movements limiting the chronic use of L-DOPA, the main pharmacological treatment of Parkinson's disease. Serotonin receptors are implicated in the development of LID and modulation of basal ganglia 5-HT1B receptors is a potential therapeutic alternative in Parkinson's disease. In the present study, we used receptor-binding autoradiography of the 5-HT1B-selective radioligand [3H]GR125743 to investigate possible contributions of changes in ligand binding of this receptor in LID in post-mortem brain specimens from Parkinson's disease patients (n=14) and control subjects (n=11), and from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys treated with saline (n=5), L-DOPA (n=4) or L-DOPA+2-methyl-6-(phenylethynyl)pyridine (MPEP) (n=5), and control monkeys (n=4). MPEP is the prototypal metabotropic glutamate 5 (mGlu5) receptor antagonist and has been shown to reduce the development of LID in these monkeys in a chronic treatment of one month. [3H]GR125743 specific binding to striatal and pallidal 5-HT1B receptors respectively were only increased in L-DOPA-treated MPTP monkeys (dyskinetic monkeys) as compared to controls, saline and L-DOPA+MPEP MPTP monkeys; dyskinesias scores correlated positively with this binding. Parkinson's disease patients with motor complications (L-DOPA-induced dyskinesias and wearing-off) had higher [3H]GR125743 specific binding compared to those without motor complications and controls in the basal ganglia. Reduction of motor complications was associated with normal striatal 5-HT1B receptors, suggesting the potential of this receptor for the management of motor complications in Parkinson's disease.
Collapse
Affiliation(s)
- Nicolas Morin
- Faculty of Pharmacy, Université Laval, Quebec City, G1K 7P4, Canada; Neuroscience Research Unit, Centre de recherche du CHU de Québec, Quebec City, G1V 4G2, Canada.
| | - Marc Morissette
- Neuroscience Research Unit, Centre de recherche du CHU de Québec, Quebec City, G1V 4G2, Canada.
| | - Laurent Grégoire
- Neuroscience Research Unit, Centre de recherche du CHU de Québec, Quebec City, G1V 4G2, Canada.
| | - Alex Rajput
- Division of Neurology, University of Saskatchewan, Royal University Hospital, Saskatoon, SK, S7N 0W8, Canada.
| | - Ali H Rajput
- Division of Neurology, University of Saskatchewan, Royal University Hospital, Saskatoon, SK, S7N 0W8, Canada.
| | - Thérèse Di Paolo
- Faculty of Pharmacy, Université Laval, Quebec City, G1K 7P4, Canada; Neuroscience Research Unit, Centre de recherche du CHU de Québec, Quebec City, G1V 4G2, Canada.
| |
Collapse
|
19
|
Jiménez-Urbieta H, Gago B, de la Riva P, Delgado-Alvarado M, Marin C, Rodriguez-Oroz MC. Dyskinesias and impulse control disorders in Parkinson's disease: From pathogenesis to potential therapeutic approaches. Neurosci Biobehav Rev 2015. [PMID: 26216865 DOI: 10.1016/j.neubiorev.2015.07.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dopaminergic treatment in Parkinson's disease (PD) reduces the severity of motor symptoms of the disease. However, its chronic use is associated with disabling motor and behavioral side effects, among which levodopa-induced dyskinesias (LID) and impulse control disorders (ICD) are the most common. The underlying mechanisms and pathological substrate of these dopaminergic complications are not fully understood. Recently, the refinement of imaging techniques and the study of the genetics and molecular bases of LID and ICD indicate that, although different, they could share some features. In addition, animal models of parkinsonism with LID have provided important knowledge about mechanisms underlying such complications. In contrast, animal models of parkinsonism and abnormal impulsivity, although useful regarding some aspects of human ICD, do not fully resemble the clinical phenotype of ICD in patients with PD, and until now have provided limited information. Studies on animal models of addiction could complement the previous models and provide some insights into the background of these behavioral complications given that ICD are regarded as behavioral addictions. Here we review the most relevant advances in relation to imaging, genetics, biochemistry and pharmacological interventions to treat LID and ICD in patients with PD and in animal models with a view to better understand the overlapping and unique maladaptations to dopaminergic therapy that are associated with LID and ICD.
Collapse
Affiliation(s)
- Haritz Jiménez-Urbieta
- Biodonostia Research Institute, 20014 San Sebastián, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Belén Gago
- Biodonostia Research Institute, 20014 San Sebastián, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | | | - Manuel Delgado-Alvarado
- Biodonostia Research Institute, 20014 San Sebastián, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Concepció Marin
- INGENIO, IRCE, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , 08036 Barcelona, Spain.
| | - María C Rodriguez-Oroz
- Biodonostia Research Institute, 20014 San Sebastián, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; University Hospital Donostia, 20014 San Sebastián, Spain; Ikerbasque (Basque Foundation for Science), 48011 Bilbao, Spain.
| |
Collapse
|
20
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
21
|
Jourdain VA, Morin N, Grégoire L, Morissette M, Di Paolo T. Changes in glutamate receptors in dyskinetic parkinsonian monkeys after unilateral subthalamotomy. J Neurosurg 2015; 123:1383-93. [PMID: 25932606 DOI: 10.3171/2014.10.jns141570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Unilateral subthalamotomy is a surgical procedure that may be used to alleviate disabling levodopa-induced dyskinesias (LIDs) in patients with Parkinson disease (PD). However, the mechanisms involved in LID remain largely unknown. The subthalamic nucleus (STN) is the sole glutamatergic nucleus within the basal ganglia, and its lesion may produce changes in glutamate receptors in various areas of the basal ganglia. The authors aimed to investigate the biochemical changes in glutamate receptors in striatal and pallidal regions of the basal ganglia after lesion of the STN in parkinsonian macaque monkeys. METHODS The authors treated 12 female ovariectomized monkeys with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce PD-like symptoms, treated 8 of these animals with 3,4-dihydroxy-l-phenylalanine (L-DOPA; levodopa) to induce LID, and performed unilateral subthalamotomy in 4 of these 8 monkeys. Four additional monkeys were treated with saline only and were used as controls. The MPTP monkeys had previously been shown to respond behaviorally to lower doses of levodopa after the STN lesion. Autoradiography of slices from postmortem brain tissues was used to visualize changes in the specific binding of striatal and pallidal ionotropic glutamate receptors (that is, of the α-amino-3-hydroxy 5-methyl-4-isoxazole propionate [AMPA] and N-methyl-d-aspartate [NMDA] NR1/NR2B subunit receptors) and of metabotropic glutamate (mGlu) receptors (that is, mGlu2/3 and mGlu5 receptors). The specific binding and distribution of glutamate receptors in the basal ganglia of the levodopa-treated, STN-lesioned MPTP monkeys were compared with those in the saline-treated control monkeys and in the saline-treated and levodopa-treated MPTP monkeys. RESULTS The autoradiographic results indicated that none of the pharmacological and surgical treatments produced changes in the specific binding of AMPA receptors in the basal ganglia. Levodopa treatment increased the specific binding of NMDA receptors in the basal ganglia. Subthalamotomy reversed these increases in the striatum, but in the globus pallidus (GP), the subthalamotomy reversed these increases only contralaterally. Levodopa treatment reversed MPTP-induced increases in mGlu2/3 receptors only in the GP. mGlu2/3 receptor-specific binding in the striatum and GP decreased bilaterally in the levodopa-treated, STN-lesioned MPTP monkeys compared with the other 3 groups. Compared with mGlu5 receptor-specific binding in the control monkeys, that of the levodopa-treated MPTP monkeys increased in the dorsal putamen and remained unchanged in the caudate nucleus and in the GP. CONCLUSIONS These results implicate glutamate receptors in the previously observed benefits of unilateral subthalamotomy to improve motor control.
Collapse
Affiliation(s)
- Vincent A Jourdain
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Nicolas Morin
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Laurent Grégoire
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec; and.,Faculty of Pharmacy, Laval University, Quebec, Canada
| |
Collapse
|
22
|
|
23
|
Morin N, Morissette M, Grégoire L, Di Paolo T. Effect of a chronic treatment with an mGlu5 receptor antagonist on brain serotonin markers in parkinsonian monkeys. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:27-38. [PMID: 25046277 DOI: 10.1016/j.pnpbp.2014.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/27/2014] [Accepted: 07/14/2014] [Indexed: 10/25/2022]
Abstract
In Parkinson's disease (PD) and l-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias (LIDs), overactivity of brain glutamate neurotransmission is documented and antiglutamatergic drugs decrease LID. Serotonin (5-HT) receptors and transporter (SERT) are also implicated in LID and we hypothesize that antiglutamatergic drugs can also regulate brain serotoninergic activity. Our aim was to investigate the long-term effect of the prototypal metabotropic glutamate 5 (mGlu5) receptor antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) with L-DOPA on basal ganglia SERT, 5-HT(1A) and 5-HT(2A) receptor levels in monkeys lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP monkeys were treated for one month with L-DOPA and developed LID while those treated with L-DOPA and MPEP (10 mg/kg) developed significantly less LID. Normal controls and saline-treated MPTP monkeys were included for biochemical analysis. The MPTP lesion and experimental treatments left unchanged striatal 5-HT concentrations. MPTP lesion induced an increase of striatal 5-HIAA concentrations similar in all MPTP monkeys as compared to controls. [(3)H]-8-OH-DPAT and [(3)H]-citalopram specific binding levels to 5-HT(1A) receptors and SERT respectively remained unchanged in the striatum and globus pallidus of all MPTP monkeys compared to controls and no difference was observed between groups of MPTP monkeys. [(3)H]-ketanserin specific binding to striatal and pallidal 5-HT2A receptors was increased in L-DOPA-treated MPTP monkeys as compared to controls, saline and L-DOPA+MPEP MPTP monkeys and no difference between the latter groups was observed; dyskinesia scores correlated positively with this binding. In conclusion, reduction of development of LID with MPEP was associated with lower striatal and pallidal 5-HT2A receptors showing that glutamate activity also affects serotoninergic markers.
Collapse
Affiliation(s)
- Nicolas Morin
- Faculty of Pharmacy, Université Laval, 1050 Avenue de la Médecine, Quebec City G1V 0A6, Canada; Neuroscience Research Unit, Centre de Recherche du CHU de Québec, 2705 Laurier Boulevard, Quebec City G1V 4G2, Canada.
| | - Marc Morissette
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, 2705 Laurier Boulevard, Quebec City G1V 4G2, Canada.
| | - Laurent Grégoire
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, 2705 Laurier Boulevard, Quebec City G1V 4G2, Canada.
| | - Thérèse Di Paolo
- Faculty of Pharmacy, Université Laval, 1050 Avenue de la Médecine, Quebec City G1V 0A6, Canada; Neuroscience Research Unit, Centre de Recherche du CHU de Québec, 2705 Laurier Boulevard, Quebec City G1V 4G2, Canada.
| |
Collapse
|
24
|
Song L, Yang X, Ma Y, Wu N, Liu Z. The CB1 cannabinoid receptor agonist reduces L-DOPA-induced motor fluctuation and ERK1/2 phosphorylation in 6-OHDA-lesioned rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:2173-9. [PMID: 25395834 PMCID: PMC4226453 DOI: 10.2147/dddt.s60944] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The dopamine precursor L-3,4-dihydroxyphenylalanine (L-DOPA) has been used as an effective drug for treating dopamine depletion-induced Parkinson’s disease (PD). However, long-term administration of L-DOPA produces motor complications. L-DOPA has also been found to modify the two key signaling cascades, protein kinase A/dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) and extracellular signal-regulated kinases 1 and 2 (ERK1/2), in striatal neurons, which are thought to play a pivotal role in forming motor complications. In the present study, we tested the possible effect of a CB1 cannabinoid receptor agonist on L-DOPA-stimulated abnormal behavioral and signaling responses in vivo. Intermittent L-DOPA administration for 3 weeks induced motor fluctuation in a rat model of PD induced by intrastriatal infusion of dopamine-depleting neurotoxin 6-hydroxydopamine (6-OHDA). A single injection of a CB1 cannabinoid receptor agonist WIN-55,212-2 had no effect on L-DOPA-induced motor fluctuation. However, chronic injections of WIN-55,212-2 significantly attenuated abnormal behavioral responses to L-DOPA in 6-OHDA-lesioned rats. Similarly, chronic injections of WIN-55,212-2 influence the L-DOPA-induced alteration of DARPP-32 and ERK1/2 phosphorylation status in striatal neurons. These data provide evidence for the active involvement of CB1 cannabinoid receptors in the regulation of L-DOPA action during PD therapy.
Collapse
Affiliation(s)
- Lu Song
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, People's Republic of China
| | - Xinxin Yang
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, People's Republic of China
| | - Yaping Ma
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, People's Republic of China
| | - Na Wu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, People's Republic of China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, People's Republic of China
| |
Collapse
|
25
|
Rascol O, Fox S, Gasparini F, Kenney C, Di Paolo T, Gomez-Mancilla B. Use of metabotropic glutamate 5-receptor antagonists for treatment of levodopa-induced dyskinesias. Parkinsonism Relat Disord 2014; 20:947-56. [DOI: 10.1016/j.parkreldis.2014.05.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/02/2014] [Accepted: 05/02/2014] [Indexed: 10/25/2022]
|
26
|
Finlay CJ, Duty S, Vernon AC. Brain morphometry and the neurobiology of levodopa-induced dyskinesias: current knowledge and future potential for translational pre-clinical neuroimaging studies. Front Neurol 2014; 5:95. [PMID: 24971074 PMCID: PMC4053925 DOI: 10.3389/fneur.2014.00095] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/29/2014] [Indexed: 11/29/2022] Open
Abstract
Dopamine replacement therapy in the form of levodopa results in a significant proportion of patients with Parkinson’s disease developing debilitating dyskinesia. This significantly complicates further treatment and negatively impacts patient quality of life. A greater understanding of the neurobiological mechanisms underlying levodopa-induced dyskinesia (LID) is therefore crucial to develop new treatments to prevent or mitigate LID. Such investigations in humans are largely confined to assessment of neurochemical and cerebrovascular blood flow changes using positron emission tomography and functional magnetic resonance imaging. However, recent evidence suggests that LID is associated with specific morphological changes in the frontal cortex and midbrain, detectable by structural MRI and voxel-based morphometry. Current human neuroimaging methods however lack sufficient resolution to reveal the biological mechanism driving these morphological changes at the cellular level. In contrast, there is a wealth of literature from well-established rodent models of LID documenting detailed post-mortem cellular and molecular measurements. The combination therefore of advanced neuroimaging methods and rodent LID models offers an exciting opportunity to bridge these currently disparate areas of research. To highlight this opportunity, in this mini-review, we provide an overview of the current clinical evidence for morphological changes in the brain associated with LID and identify potential cellular mechanisms as suggested from human and animal studies. We then suggest a framework for combining small animal MRI imaging with rodent models of LID, which may provide important mechanistic insights into the neurobiology of LID.
Collapse
Affiliation(s)
- Clare J Finlay
- Wolfson Centre for Age-related Diseases, King's College London , London , UK
| | - Susan Duty
- Wolfson Centre for Age-related Diseases, King's College London , London , UK
| | - Anthony C Vernon
- Department of Neuroscience, James Black Centre, Institute of Psychiatry, King's College London , London , UK
| |
Collapse
|
27
|
Modeling dyskinesia in animal models of Parkinson disease. Exp Neurol 2014; 256:105-16. [DOI: 10.1016/j.expneurol.2013.01.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/12/2013] [Accepted: 01/21/2013] [Indexed: 01/23/2023]
|
28
|
Phenotypic screening of the ToxCast chemical library to classify toxic and therapeutic mechanisms. Nat Biotechnol 2014; 32:583-91. [PMID: 24837663 DOI: 10.1038/nbt.2914] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 04/23/2014] [Indexed: 11/08/2022]
Abstract
Addressing the safety aspects of drugs and environmental chemicals has historically been undertaken through animal testing. However, the quantity of chemicals in need of assessment and the challenges of species extrapolation require the development of alternative approaches. Our approach, the US Environmental Protection Agency's ToxCast program, utilizes a large suite of in vitro and model organism assays to interrogate important chemical libraries and computationally analyze bioactivity profiles. Here we evaluated one component of the ToxCast program, the use of primary human cell systems, by screening for chemicals that disrupt physiologically important pathways. Chemical-response signatures for 87 endpoints covering molecular functions relevant to toxic and therapeutic pathways were generated in eight cell systems for 641 environmental chemicals and 135 reference pharmaceuticals and failed drugs. Computational clustering of the profiling data provided insights into the polypharmacology and potential off-target effects for many chemicals that have limited or no toxicity information. The endpoints measured can be closely linked to in vivo outcomes, such as the upregulation of tissue factor in endothelial cell systems by compounds linked to the risk of thrombosis in vivo. Our results demonstrate that assaying complex biological pathways in primary human cells can identify potential chemical targets, toxicological liabilities and mechanisms useful for elucidating adverse outcome pathways.
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
|
Cui G, Yang X, Wang X, Zhang Z, Yue X, Shi H, Shen X. Ranitidine reduced levodopa-induced dyskinesia in a rat model of Parkinson's disease. Neuropsychiatr Dis Treat 2014; 10:39-46. [PMID: 24379672 PMCID: PMC3872142 DOI: 10.2147/ndt.s54782] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Chronic administration of levodopa in Parkinson's disease leads to debilitating involuntary movements, termed levodopa-induced dyskinesia (LID). The pathogenesis of LID is poorly understood. Previous research has shown that histamine H2 receptors are highly expressed in the input (striatum) and output (globus pallidus, substantia nigra) regions of the basal ganglia, particularly in the GABAergic striatopallidal and striatonigral pathways. Therefore, a histamine H2 receptor antagonist could be used to reduce LID. In the present work, we investigated whether ranitidine has the potential to diminish LID in rats with dyskinesia and explored the underlying mechanisms involved. METHODS A rat model of PD was induced by 6-hydroxydopamine. Valid PD rats were then treated with levodopa (25 mg/kg, intraperitoneally) and benserazide (12.5 mg/kg, intraperitoneally) for 21 days to induce a rat model of LID. The acute and chronic effects of administration of ranitidine at different doses (5 mg/kg, 10 mg/kg, and 20 mg/kg) on abnormal involuntary movements, levodopa-induced rotations, and the forelimb adjusting steps test were investigated in LID rats. The chronic effect of ranitidine (10 mg/kg) on the expression of Arc and proenkephalin was also evaluated. RESULTS Levodopa elicited increased dyskinesia in PD rats. Acute ranitidine treatment had no effect on LID, but chronic ranitidine administration (10 mg/kg, 20 mg/kg) reduced LID in rats with dyskinesia. Importantly, levodopa-induced rotations were not affected by chronic treatment with ranitidine. In addition, chronic ranitidine (10 mg/kg, 20 mg/kg) significantly improved stepping of the lesioned forepaw. Real-time polymerase chain reaction showed that Arc and proenkephalin levels were reduced by chronic ranitidine (10 mg/kg) in dyskinetic rats. CONCLUSION These data indicate that ranitidine is a good adjunct for reducing LID in rats with dyskinesia. Inhibition of dopamine D1-mediated activation in the medium spiny neurons may account for the antidyskinetic effects of ranitidine in rats with dyskinesia.
Collapse
Affiliation(s)
- Guiyun Cui
- Department of Neurology, Jiangsu, People's Republic of China ; Department of Neurology, Jiangsu, People's Republic of China
| | - Xinxin Yang
- Department of Neurology, Jiangsu, People's Republic of China ; Department of Neurology, Jiangsu, People's Republic of China
| | - Xiaoying Wang
- Department of Ultrasound, the Affiliated Hospital of Xuzhou Medical College, Jiangsu, People's Republic of China ; Department of Neurology, Jiangsu, People's Republic of China
| | - Zunsheng Zhang
- Department of Neurology, Jiangsu, People's Republic of China
| | - Xuanye Yue
- Department of Neurology, Jiangsu, People's Republic of China
| | - Hongjuan Shi
- Department of Neurology, Jiangsu, People's Republic of China
| | - Xia Shen
- Department of Neurology, Jiangsu, People's Republic of China
| |
Collapse
|
31
|
Kintz N, Petzinger GM, Akopian G, Ptasnik S, Williams C, Jakowec MW, Walsh JP. Exercise modifies α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor expression in striatopallidal neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned mouse. J Neurosci Res 2013; 91:1492-507. [PMID: 23918451 DOI: 10.1002/jnr.23260] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/26/2013] [Accepted: 05/16/2013] [Indexed: 12/11/2022]
Abstract
The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic-acid-type glutamate receptor (AMPAR) plays a critical role in modulating experience-dependent neuroplasticity, and alterations in AMPAR expression may underlie synaptic dysfunction and disease pathophysiology. Using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of dopamine (DA) depletion, our previous work showed exercise increases total GluA2 subunit expression and the contribution of GluA2-containing channels in MPTP mice. The purpose of this study was to determine whether exercise-dependent changes in AMPAR expression after MPTP are specific to the striatopallidal (D2 R) or striatonigral (D1 R) medium spiny neuron (MSN) striatal projection pathways. Drd2 -eGFP-BAC transgenic mice were used to delineate differences in AMPAR expression between striatal D2 R-MSNs and D1 R-MSNs. Striatal AMPAR expression was assessed by immunohistochemical (IHC) staining, Western immunoblotting (WB) of preparations enriched for postsynaptic density (PSD), and alterations in the current-voltage relationship of MSNs. We found DA depletion results in the emergence of GluA2-lacking AMPARs selectively in striatopallidal D2 R-MSNs and that exercise reverses this effect in MPTP mice. Exercise-induced changes in AMPAR channels observed after DA depletion were associated with alterations in GluA1 and GluA2 subunit expression in postsynaptic protein, D2 R-MSN cell surface expression, and restoration of corticostriatal plasticity. Mechanisms regulating experience-dependent changes in AMPAR expression may provide innovative therapeutic targets to increase the efficacy of treatments for basal ganglia disorders, including Parkinson's disease.
Collapse
Affiliation(s)
- N Kintz
- The George and MaryLou Boone Center for Parkinson's Disease Research, Department of Neurology, University of Southern California, Los Angeles, California
| | | | | | | | | | | | | |
Collapse
|
32
|
Morin N, Morissette M, Grégoire L, Gomez-Mancilla B, Gasparini F, Di Paolo T. Chronic treatment with MPEP, an mGlu5 receptor antagonist, normalizes basal ganglia glutamate neurotransmission in L-DOPA-treated parkinsonian monkeys. Neuropharmacology 2013; 73:216-31. [PMID: 23756168 DOI: 10.1016/j.neuropharm.2013.05.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 05/17/2013] [Accepted: 05/18/2013] [Indexed: 10/26/2022]
Abstract
Metabotropic glutamate 5 (mGlu5) receptor antagonists reduce L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias (LID) in Parkinson's disease (PD). The aim of this study was to investigate the long-term effect of the prototypal mGlu5 receptor antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) on glutamate receptors known to be involved in the development of LID in the de novo chronic treatment of monkeys lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP monkeys were treated for one month with L-DOPA and developed dyskinesias while those treated with L-DOPA and MPEP (10 mg/kg) developed significantly less. Normal control and saline-treated MPTP monkeys were also included. All MPTP monkeys were extensively and similarly denervated. The basal ganglia [(3)H]ABP688 specific binding (mGlu5 receptors) was elevated in L-DOPA-treated MPTP monkeys compared to controls but not in those treated with L-DOPA and MPEP; dyskinesia scores of these monkeys correlated positively with their [(3)H]ABP688 specific binding. Striatal density (B(max)) of [(3)H]ABP688 specific binding increased in L-DOPA-treated MPTP monkeys compared to other groups and affinity (Kd) remained unchanged. Striatal mGlu5 receptor mRNA remained unchanged following treatments. Elevated basal ganglia specific binding of [(3)H]Ro 25-6981 (NMDA NR1/NR2B receptors), [(3)H]Ro 48-8587 (AMPA receptors) but not [(3)H]CGP-39653 (NMDA NR1/NR2A receptors) was observed only in L-DOPA-treated MPTP monkeys; dyskinesias scores correlated with binding. By contrast, basal ganglia [(3)H]LY341495 specific binding (mGlu2/3 receptors) decreased in L-DOPA-treated MPTP monkeys compared to controls, saline and L-DOPA + MPEP treated MPTP monkeys; dyskinesias scores correlated negatively with this binding. Hence, chronic MPEP treatment reduces the development of LID and is associated with a normalization of glutamate neurotransmission.
Collapse
Affiliation(s)
- Nicolas Morin
- Neuroscience Research Unit, Laval University Medical Center (CHUQ), Quebec, QC, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Yang X, Wu N, Song L, Liu Z. Intrastriatal injections of KN-93 ameliorates levodopa-induced dyskinesia in a rat model of Parkinson's disease. Neuropsychiatr Dis Treat 2013; 9:1213-20. [PMID: 23983471 PMCID: PMC3751461 DOI: 10.2147/ndt.s45422] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Levodopa remains the most effective drug for the treatment of Parkinson's disease (PD). However, long-term levodopa treatment is associated with the emergence of levodopa-induced dyskinesia (LID), which has hampered its use for PD treatment. The mechanisms of LID are only partially understood. A previous study showed that KN-93, a Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) inhibitor, could be used to ameliorate LID in rats. However, the precise mechanisms by which KN-93 acts as an antidyskinetic are not fully understood. METHODS In the present study, a rat model of PD was induced by 6-hydroxydopamine (OHDA) injections. Then, the successfully lesioned rats were intrastriatally administered with a different dose of KN-93 (1 μg, 2 μg, or 5 μg) prior to levodopa treatment. Abnormal involuntary movements (AIMs) scores and apomorphine-induced rotations were measured in PD rats. Phosphorylated levels of GluR1 at Serine-845 (pGluR1S845) levels were determined by western blot. Arc and Penk levels were measured by real-time polymerase chain reaction (PCR). RESULTS We found that both 2 μg and 5 μg KN-93 treatment lowered AIMs scores in levodopa priming PD rats without affecting the antiparkinsonian effect of levodopa. In agreement with behavioral analysis, KN-93 treatment (2 μg) reduced pGluR1S845 levels in PD rats. Moreover, KN-93 treatment (2 μg) reduced the expression of Gad1 and Nur77 in PD rats. CONCLUSION These data indicated that intrastriatal injections of KN-93 were beneficial in reducing the expression of LID by lowering the expression of pGluR1S845 via suppressing the activation of CaMKII in PD rats. Decreased expression of pGluR1S845 further reduced the expression of Gad1 and Nur77 in PD rats.
Collapse
Affiliation(s)
- Xinxin Yang
- Department of Neurology, Xinhua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
34
|
Huot P, Johnston TH, Koprich JB, Fox SH, Brotchie JM. The pharmacology of L-DOPA-induced dyskinesia in Parkinson's disease. Pharmacol Rev 2013; 65:171-222. [PMID: 23319549 DOI: 10.1124/pr.111.005678] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
L-3,4-Dihydroxyphenylalanine (L-DOPA) remains the most effective symptomatic treatment of Parkinson's disease (PD). However, long-term administration of L-DOPA is marred by the emergence of abnormal involuntary movements, i.e., L-DOPA-induced dyskinesia (LID). Years of intensive research have yielded significant progress in the quest to elucidate the mechanisms leading to the development and expression of dyskinesia and maintenance of the dyskinetic state, but the search for a complete understanding is still ongoing. Herein, we summarize the current knowledge of the pharmacology of LID in PD. Specifically, we review evidence gathered from postmortem and pharmacological studies, both preclinical and clinical, and discuss the involvement of dopaminergic and nondopaminergic systems, including glutamatergic, opioid, serotonergic, γ-aminobutyric acid (GABA)-ergic, adenosine, cannabinoid, adrenergic, histaminergic, and cholinergic systems. Moreover, we discuss changes occurring in transcription factors, intracellular signaling, and gene expression in the dyskinetic phenotype. Inasmuch as a multitude of neurotransmitters and receptors play a role in the etiology of dyskinesia, we propose that to optimally alleviate this motor complication, it may be necessary to develop combined treatment approaches that will target simultaneously more than one neurotransmitter system. This could be achieved via three ways as follows: 1) by developing compounds that will interact simultaneously to a multitude of receptors with the required agonist/antagonist effect at each target, 2) by targeting intracellular signaling cascades where the signals mediated by multiple receptors converge, and/or 3) to regulate gene expression in a manner that has effects on signaling by multiple pathways.
Collapse
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
35
|
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
|
36
|
Yang X, Chen Y, Hong X, Wu N, Song L, Yuan W, Liu Z. Levodopa/benserazide microspheres reduced levodopa-induced dyskinesia by downregulating phosphorylated GluR1 expression in 6-OHDA-lesioned rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2012. [PMID: 23185117 PMCID: PMC3506046 DOI: 10.2147/dddt.s38008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Levodopa is the gold standard in the treatment of Parkinson’s disease (PD). However, long-term levodopa replacement therapy is accompanied by abnormal involuntary movements (AIMs), known as levodopa-induced dyskinesia (LID). Until now, the precise mechanisms of LID were only partially understood. Previous studies have shown that continuous dopamine stimulation was helpful in reducing the expression of LID. In addition to dopamine D1 receptor, glutamatergic receptors such as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor also contribute to the expression of LID. The current authors have previously reported that levodopa/benserazide-loaded microspheres could ameliorate the expression of LID by reducing the protein kinase A signaling pathway in dyskinetic rats. However, whether AMPA receptor is involved in the mechanism by which levodopa/benserazide-loaded microspheres ameliorate the expression of LID in dyskinetic rats was unknown. Methods In the present study, as reported previously, levodopa and benserazide were loaded by poly(lactic-co-glycolic acid) microspheres, which can release levodopa and benserazide in a sustained manner. 6-Hydroxydopamine was injected into the right medial forebrain bundle to produce a rat model of PD. Then valid PD rats were treated with levodopa plus benserazide for 3 weeks to induce a rat model of LID. Dyskinetic rats were treated with levodopa/beserazide-loaded microspheres containing levodopa (6 mg/kg) plus benserazide (15 mg/kg) or same dose of levodopa plus benserazide. Abnormal involuntary movements were measured in rats on days 1, 5, 10, 15, and 20 during the treatment. The levels of GluR1 at serine-831 (pGluR1S831) and serine-845 (pGluR1S845) were determined by Western blot. Arc and proenkephalin (Penk) levels were measured by real-time polymerase chain reaction. Results Three-week levodopa plus benserazide treatment induced dyskinesia in PD rats. Levodopa/benserazide-loaded microsphere-treated dyskinetic rats showed lower AIM scores than levodopa plus benserazide-treated dyskinetic rats. Microsphere treatment downregulated the phosphrylated levels of pGluR1S831 and pGluR1S845 in the striatum of dyskinetic rats. In addition, microsphere treatment reduced the levels of Arc and Penk. Conclusion These data indicated that levodopa/benserazide-loaded microspheres could be used to ameliorate the expression of LID by reducing the expression of pGluR1S831 and pGluR1S845 as well as Arc and Penk.
Collapse
Affiliation(s)
- Xinxin Yang
- Department of Neurology, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Steece-Collier K, Rademacher DJ, Soderstrom K. Anatomy of Graft-induced Dyskinesias: Circuit Remodeling in the Parkinsonian Striatum. ACTA ACUST UNITED AC 2012; 2:15-30. [PMID: 22712056 DOI: 10.1016/j.baga.2012.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The goal of researchers and clinicians interested in re-instituting cell based therapies for PD is to develop an effective and safe surgical approach to replace dopamine (DA) in individuals suffering from Parkinson's disease (PD). Worldwide clinical trials involving transplantation of embryonic DA neurons into individuals with PD have been discontinued because of the often devastating post-surgical side-effect known as graft-induced dyskinesia (GID). There have been many review articles published in recent years on this subject. There has been a tendency to promote single factors in the cause of GID. In this review, we contrast the pros and cons of multiple factors that have been suggested from clinical and/or preclinical observations, as well as novel factors not yet studied that may be involved with GID. It is our intention to provide a platform that might be instrumental in examining how individual factors that correlate with GID and/or striatal pathology might interact to give rise to dysfunctional circuit remodeling and aberrant motor output.
Collapse
Affiliation(s)
- Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503
| | | | | |
Collapse
|
38
|
Blandini F, Armentero MT. New pharmacological avenues for the treatment of L-DOPA-induced dyskinesias in Parkinson's disease: targeting glutamate and adenosine receptors. Expert Opin Investig Drugs 2012; 21:153-68. [PMID: 22233485 DOI: 10.1517/13543784.2012.651457] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) therapy is still centered on the use of L-3,4-dihydroxyphenylalanine (L-DOPA), which is hampered by numerous side effects, including abnormal involuntary movements known as L-DOPA-induced dyskinesias (LIDs). LIDs are the result of pre- and postsynaptic changes at the corticostriatal level, induced by chronic and pulsatile stimulation of striatal dopaminergic receptors. These changes impact on synaptic plasticity and involve also selected, nondopaminergic receptors expressed by striatal projection neurons. AREAS COVERED Among nondopaminergic receptors, glutamate receptors - NMDA and mGluR5 subtypes in particular - and adenosine A(2A) receptors are those most likely involved in LIDs. The aim of the present review is to summarize results of studies undertaken with specific antagonists of these receptors, first conducted in animal models of LIDs, which in selected cases have been translated into clinical trials. EXPERT OPINION Selected antagonists of glutamate and adenosine receptors have been proposed as anti-dyskinetic agents. Promising results have been obtained in preclinical investigations and in initial clinical trials, but long-term safety, tolerability and efficacy studies in patients are still required. The current development of novel antagonists, including tools able to act on receptor mosaics, may provide innovative tools for LIDs management in the next future.
Collapse
Affiliation(s)
- Fabio Blandini
- IRCCS National Neurological Institute C. Mondino, Interdepartmental Research Center for Parkinson's Disease, Via Mondino 2, 27100 Pavia, Italy.
| | | |
Collapse
|
39
|
Sgambato-Faure V, Cenci MA. Glutamatergic mechanisms in the dyskinesias induced by pharmacological dopamine replacement and deep brain stimulation for the treatment of Parkinson's disease. Prog Neurobiol 2012; 96:69-86. [DOI: 10.1016/j.pneurobio.2011.10.005] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 10/25/2011] [Accepted: 10/27/2011] [Indexed: 12/13/2022]
|
40
|
Huang YZ, Rothwell JC, Lu CS, Chuang WL, Chen RS. Abnormal bidirectional plasticity-like effects in Parkinson's disease. Brain 2011; 134:2312-20. [DOI: 10.1093/brain/awr158] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
41
|
Dupre KB, Ostock CY, Jaunarajs KLE, Button T, Savage LM, Wolf W, Bishop C. Local modulation of striatal glutamate efflux by serotonin 1A receptor stimulation in dyskinetic, hemiparkinsonian rats. Exp Neurol 2011; 229:288-99. [PMID: 21352823 PMCID: PMC3100430 DOI: 10.1016/j.expneurol.2011.02.012] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/10/2011] [Accepted: 02/15/2011] [Indexed: 10/18/2022]
Abstract
Serotonin 1A receptor (5-HT(1A)R) agonists reduce both L-DOPA- and D1 receptor (D1R) agonist-mediated dyskinesia, but their anti-dyskinetic mechanism of action is not fully understood. Given that 5-HT(1A)R stimulation reduces glutamatergic neurotransmission in the dopamine-depleted striatum, 5-HT(1A)R agonists may diminish dyskinesia in part through modulation of pro-dyskinetic striatal glutamate levels. To test this, rats with unilateral medial forebrain bundle dopamine or sham lesions were primed with L-DOPA (12 mg/kg+benserazide, 15 mg/kg, sc) or the D1R agonist SKF81297 (0.8 mg/kg, sc) until abnormal involuntary movements (AIMs) stabilized. On subsequent test days, rats were treated with vehicle or the 5-HT(1A)R agonist ±8-OH-DPAT (1.0 mg/kg, sc), followed by L-DOPA or SKF81297, or intrastriatal ±8-OH-DPAT (7.5 or 15 mM), followed by L-DOPA. In some cases, the 5-HT(1A)R antagonist WAY100635 was employed to determine receptor-specific effects. In vivo microdialysis was used to collect striatal samples for analysis of extracellular glutamate levels during AIMs assessment. Systemic and striatal ±8-OH-DPAT attenuated L-DOPA-induced dyskinesia and striatal glutamate efflux while WAY100635 reversed ±8-OH-DPAT's effects. Interestingly, systemic ±8-OH-DPAT diminished D1R-mediated AIMs without affecting glutamate. These findings indicate a novel anti-dyskinetic mechanism of action for 5-HT(1A)R agonists with implications for the improved treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Kristin B. Dupre
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| | - Corinne Y. Ostock
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| | - Karen L. Eskow Jaunarajs
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| | - Thomas Button
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| | - Lisa M. Savage
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| | - William Wolf
- Research Service, Hines VA Hospital Hines, IL, 60141 and Anatomy & Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University (State University of New York at Binghamton), Binghamton, NY 13902-6000, USA
| |
Collapse
|