1
|
Mews P, Van der Zee Y, Gurung A, Estill M, Futamura R, Kronman H, Ramakrishnan A, Ryan M, Reyes AA, Garcia BA, Browne CJ, Sidoli S, Shen L, Nestler EJ. Cell type-specific epigenetic priming of gene expression in nucleus accumbens by cocaine. SCIENCE ADVANCES 2024; 10:eado3514. [PMID: 39365860 PMCID: PMC11451531 DOI: 10.1126/sciadv.ado3514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024]
Abstract
A hallmark of addiction is the ability of drugs of abuse to trigger relapse after periods of prolonged abstinence. Here, we describe an epigenetic mechanism whereby chronic cocaine exposure causes lasting chromatin and downstream transcriptional modifications in the nucleus accumbens (NAc), a critical brain region controlling motivation. We link prolonged withdrawal from cocaine to the depletion of the histone variant H2A.Z, coupled with increased genome accessibility and latent priming of gene transcription, in D1 dopamine receptor-expressing medium spiny neurons (D1 MSNs) that relate to aberrant gene expression upon drug relapse. The histone chaperone ANP32E removes H2A.Z from chromatin, and we demonstrate that D1 MSN-selective Anp32e knockdown prevents cocaine-induced H2A.Z depletion and blocks cocaine's rewarding actions. By contrast, very different effects of cocaine exposure, withdrawal, and relapse were found for D2 MSNs. These findings establish histone variant exchange as an important mechanism and clinical target engaged by drugs of abuse to corrupt brain function and behavior.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Van der Zee
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashik Gurung
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hope Kronman
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meagan Ryan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abner A. Reyes
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medticine, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Sáez M, Keifman E, Alberquilla S, Coll C, Reig R, Murer MG, Moratalla R. D2 dopamine receptors and the striatopallidal pathway modulate L-DOPA-induced dyskinesia in the mouse. Neurobiol Dis 2023; 186:106278. [PMID: 37683958 DOI: 10.1016/j.nbd.2023.106278] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
L-DOPA-induced dyskinesia (LID) remains a major complication of Parkinson's disease management for which better therapies are necessary. The contribution of the striatonigral direct pathway to LID is widely acknowledged but whether the striatopallidal pathway is involved remains debated. Selective optogenetic stimulation of striatonigral axon terminals induces dyskinesia in mice rendered hemiparkinsonian with the toxin 6-hydroxydopamine (6-OHDA). Here we show that optogenetically-induced dyskinesia is increased by the D2-type dopamine receptor agonist quinpirole. Although the quinpirole effect may be mediated by D2 receptor stimulation in striatopallidal neurons, alternative mechanisms may be responsible as well. To selectively modulate the striatopallidal pathway, we selectively expressed channelrhodopsin-2 (ChR2) in D2 receptor expressing neurons by crossing D2-Cre and ChR2-flox mice. The animals were rendered hemiparkinsonian and implanted with an optic fiber at the ipsilateral external globus pallidus (GPe). Stimulation of ChR2 at striatopallidal axon terminals reduced LID and also general motility during the off L-DOPA state, without modifying the pro-motor effect of low doses of L-DOPA producing mild or no dyskinesia. Overall, the present study shows that D2-type dopamine receptors and the striatopallidal pathway modulate dyskinesia and suggest that targeting striatopallidal axon terminals at the GPe may have therapeutic potential in the management of LID.
Collapse
Affiliation(s)
- María Sáez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain; Instituto de Neurociencias UMH-CSIC, San Juan de Alicante, Alicante 03550, Spain
| | - Ettel Keifman
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Argentina; Universidad de Buenos Aires and CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), 2155 Paraguay St, Buenos Aires 1121, Argentina
| | - Samuel Alberquilla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Camila Coll
- Universidad de Buenos Aires and CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), 2155 Paraguay St, Buenos Aires 1121, Argentina
| | - Ramón Reig
- Instituto de Neurociencias UMH-CSIC, San Juan de Alicante, Alicante 03550, Spain
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Argentina; Universidad de Buenos Aires and CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), 2155 Paraguay St, Buenos Aires 1121, Argentina.
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Liu K, Song M, Gao S, Yao L, Zhang L, Feng J, Wang L, Gao R, Wang Y. The Dynamics of Dopamine D 2 Receptor-Expressing Striatal Neurons and the Downstream Circuit Underlying L-Dopa-Induced Dyskinesia in Rats. Neurosci Bull 2023; 39:1411-1425. [PMID: 37022638 PMCID: PMC10465438 DOI: 10.1007/s12264-023-01054-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 04/07/2023] Open
Abstract
L-dopa (l-3,4-dihydroxyphenylalanine)-induced dyskinesia (LID) is a debilitating complication of dopamine replacement therapy for Parkinson's disease. The potential contribution of striatal D2 receptor (D2R)-positive neurons and downstream circuits in the pathophysiology of LID remains unclear. In this study, we investigated the role of striatal D2R+ neurons and downstream globus pallidus externa (GPe) neurons in a rat model of LID. Intrastriatal administration of raclopride, a D2R antagonist, significantly inhibited dyskinetic behavior, while intrastriatal administration of pramipexole, a D2-like receptor agonist, yielded aggravation of dyskinesia in LID rats. Fiber photometry revealed the overinhibition of striatal D2R+ neurons and hyperactivity of downstream GPe neurons during the dyskinetic phase of LID rats. In contrast, the striatal D2R+ neurons showed intermittent synchronized overactivity in the decay phase of dyskinesia. Consistent with the above findings, optogenetic activation of striatal D2R+ neurons or their projections in the GPe was adequate to suppress most of the dyskinetic behaviors of LID rats. Our data demonstrate that the aberrant activity of striatal D2R+ neurons and downstream GPe neurons is a decisive mechanism mediating dyskinetic symptoms in LID rats.
Collapse
Affiliation(s)
- Kuncheng Liu
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
- Department of Clinical Medicine, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Miaomiao Song
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Shasha Gao
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Lu Yao
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Li Zhang
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Jie Feng
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China
| | - Ling Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710004, China
| | - Rui Gao
- Department of Medical Imaging and Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| | - Yong Wang
- Department of Physiology and Pathophysiology and Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
4
|
Zell L, Bretl A, Temml V, Schuster D. Dopamine Receptor Ligand Selectivity-An In Silico/In Vitro Insight. Biomedicines 2023; 11:1468. [PMID: 37239139 PMCID: PMC10216180 DOI: 10.3390/biomedicines11051468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Different dopamine receptor (DR) subtypes are involved in pathophysiological conditions such as Parkinson's Disease (PD), schizophrenia and depression. While many DR-targeting drugs have been approved by the U.S. Food and Drug Administration (FDA), only a very small number are truly selective for one of the DR subtypes. Additionally, most of them show promiscuous activity at related G-protein coupled receptors, thus suffering from diverse side-effect profiles. Multiple studies have shown that combined in silico/in vitro approaches are a valuable contribution to drug discovery processes. They can also be applied to divulge the mechanisms behind ligand selectivity. In this study, novel DR ligands were investigated in vitro to assess binding affinities at different DR subtypes. Thus, nine D2R/D3R-selective ligands (micro- to nanomolar binding affinities, D3R-selective profile) were successfully identified. The most promising ligand exerted nanomolar D3R activity (Ki = 2.3 nM) with 263.7-fold D2R/D3R selectivity. Subsequently, ligand selectivity was rationalized in silico based on ligand interaction with a secondary binding pocket, supporting the selectivity data determined in vitro. The developed workflow and identified ligands could aid in the further understanding of the structural motifs responsible for DR subtype selectivity, thus benefitting drug development in D2R/D3R-associated pathologies such as PD.
Collapse
Affiliation(s)
| | | | | | - Daniela Schuster
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria; (L.Z.); (A.B.); (V.T.)
| |
Collapse
|
5
|
Ma YF, Lin YA, Huang CL, Hsu CC, Wang S, Yeh SR, Tsai YC. Lactiplantibacillus plantarum PS128 Alleviates Exaggerated Cortical Beta Oscillations and Motor Deficits in the 6-Hydroxydopamine Rat Model of Parkinson's Disease. Probiotics Antimicrob Proteins 2023; 15:312-325. [PMID: 34449056 DOI: 10.1007/s12602-021-09828-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by midbrain dopaminergic neuronal loss and subsequent physical impairments. Levodopa manages symptoms best, while deep brain stimulation (DBS) is effective for advanced PD patients; however, side effects occur with the diminishing therapeutic window. Recently, Lactiplantibacillus plantarum PS128 (PS128) was found to elevate dopamine levels in rodent brains, suggesting its potential to prevent PD. Here, the therapeutic efficacy of PS128 was examined in the 6-hydroxydopamine rat PD model. Suppression of the power spectral density of beta oscillations (beta PSD) in the primary motor cortex (M1) was recorded as the indicator of disease progression. We found that 6 weeks of daily PS128 supplementation suppressed M1 beta PSD as well as did levodopa and DBS. Long-term normalization of M1 beta PSD was found in PS128-fed rats, whereas levodopa and DBS showed only temporal effects. PS128 + levodopa and PS128 + DBS exhibited better therapeutic effects than did levodopa + DBS or either alone. Significantly improved motor functions in PS128-fed rats were correlated with normalization of M1 beta PSD. Brain tissue analyses further demonstrated the role of PS128 in dopaminergic neuroprotection and the enhanced availability of neurotransmitters. These findings suggest that psychobiotic PS128 might be used alongside conventional therapies to treat PD patients.
Collapse
Affiliation(s)
- Yi-Fan Ma
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Microbiome Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yi-An Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 300, Taiwan
- EzInstrument Technology Co., Ltd., Hsinchu, 300, Taiwan
| | - Chin-Lin Huang
- Microbiome Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Bened Biomedical Co., Ltd., Taipei, 104, Taiwan
| | | | - Sabrina Wang
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Shih-Rung Yeh
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Ying-Chieh Tsai
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Microbiome Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
6
|
Szalisznyó K, Silverstein DN. Why Does Tardive Dyskinesia Have Oro-facial Predominance? A Network Analysis. Brain Topogr 2023; 36:99-105. [PMID: 36592263 PMCID: PMC9834360 DOI: 10.1007/s10548-022-00931-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/27/2022] [Indexed: 01/03/2023]
Abstract
Tardive dyskinesia is a involuntary hyperkinetic disorder which usually occurs in older patients after long-term treatment with antipsychotic drugs. These dyskinesias are mostly irreversible and are frequently expressed in the tongue, cheeks, mandible, perioral area and other regions of the face. In this theoretical study we asked the question, why does tardive dyskinesia often have orofacial predominance? What might be the underlying neural network structure which contributes to this propensity? Graph analysis of high-level cortico-striato-thalamo-cortical network structure suggests a connectivity bottleneck. The number of walks of different lengths from the substantia nigra pars reticulata (SNr) to other vertices, as well as the returning cycles are the lowest in the network, which may indicate a higher damage susceptibility of this node. Analysis was also performed on published data from a recent high resolution histological study on cortico-striato-thalamo-cortical networks in rodents. Finer network partitioning and adjacency matrices demonstrated that the SNr has a heterogeneous connectivity structure and the number of local walks from nodes neighboring orofacial neural representation is higher, indicating possible early compensatory escape routes. However, with more extensive SNr damage the larger circuit compensation might be limited. This area of inquiry is important for future research, because identifying key vulnerable structures may provide more targeted therapeutical interventions.
Collapse
Affiliation(s)
- Krisztina Szalisznyó
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala University Hospital, 75185 Uppsala, Sweden
- Computational Sciences Department, Theoretical Neuroscience and Complex Systems Research Group, Wigner Research Centre for Physics, Budapest, 1121 Hungary
| | | |
Collapse
|
7
|
Targeting G Protein-Coupled Receptors in the Treatment of Parkinson's Disease. J Mol Biol 2022:167927. [PMID: 36563742 DOI: 10.1016/j.jmb.2022.167927] [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: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized in part by the deterioration of dopaminergic neurons which leads to motor impairment. Although there is no cure for PD, the motor symptoms can be treated using dopamine replacement therapies including the dopamine precursor L-DOPA, which has been in use since the 1960s. However, neurodegeneration in PD is not limited to dopaminergic neurons, and many patients experience non-motor symptoms including cognitive impairment or neuropsychiatric disturbances, for which there are limited treatment options. Moreover, there are currently no treatments able to alter the progression of neurodegeneration. There are many therapeutic strategies being investigated for PD, including alternatives to L-DOPA for the treatment of motor impairment, symptomatic treatments for non-motor symptoms, and neuroprotective or disease-modifying agents. G protein-coupled receptors (GPCRs), which include the dopamine receptors, are highly druggable cell surface proteins which can regulate numerous intracellular signaling pathways and thereby modulate the function of neuronal circuits affected by PD. This review will describe the treatment strategies being investigated for PD that target GPCRs and their downstream signaling mechanisms. First, we discuss new developments in dopaminergic agents for alleviating PD motor impairment, the role of dopamine receptors in L-DOPA induced dyskinesia, as well as agents targeting non-dopamine GPCRs which could augment or replace traditional dopaminergic treatments. We then discuss GPCRs as prospective treatments for neuropsychiatric and cognitive symptoms in PD. Finally, we discuss the evidence pertaining to ghrelin receptors, β-adrenergic receptors, angiotensin receptors and glucagon-like peptide 1 receptors, which have been proposed as disease modifying targets with potential neuroprotective effects in PD.
Collapse
|
8
|
Paz RM, Stahl AM, Rela L, Murer MG, Tubert C. D1/D5 Inverse Agonists Restore Striatal Cholinergic Interneuron Physiology in Dyskinetic Mice. Mov Disord 2022; 37:1693-1706. [PMID: 35535012 DOI: 10.1002/mds.29055] [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: 11/16/2021] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND In advanced stages of Parkinson's disease (PD), dyskinesia and motor fluctuations become seriously debilitating and therapeutic options become scarce. Aberrant activity of striatal cholinergic interneurons (SCIN) has been shown to be critical to PD and dyskinesia, but the systemic administration of cholinergic medications can exacerbate extrastriatal-related symptoms. Thus, targeting the mechanisms causing pathological SCIN activity in severe PD with motor fluctuations and dyskinesia is a promising therapeutic alternative. METHODS We used ex vivo electrophysiological recordings combined with pharmacology to study the alterations in intracellular signaling that contribute to the altered SCIN physiology observed in the 6-hydroxydopamine mouse model of PD treated with levodopa. RESULTS The altered phenotypes of SCIN of parkinsonian mice during the "off levodopa" state resulting from aberrant Kir/leak and Kv1.3 currents can be rapidly reverted by acute inhibition of cAMP-ERK1/2 signaling. Inverse agonists that inhibit the ligand-independent activity of D5 receptors, like clozapine, restore Kv1.3 and Kir/leak currents and SCIN normal physiology in dyskinetic mice. CONCLUSION Our work unravels a signaling pathway involved in the dysregulation of membrane currents causing SCIN hyperexcitability and burst-pause activity in parkinsonian mice treated with levodopa (l-dopa). These changes persist during off-medication periods due to tonic mechanisms that can be acutely reversed by pharmacological interventions. Thus, targeting the D5-cAMP-ERK1/2 signaling pathway selectively in SCIN may have therapeutic effects in PD and dyskinesia by restoring the normal SCIN function. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rodrigo Manuel Paz
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Agostina Mónica Stahl
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Lorena Rela
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Cecilia Tubert
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| |
Collapse
|
9
|
Chagraoui A, Di Giovanni G, De Deurwaerdère P. Neurobiological and Pharmacological Perspectives of D3 Receptors in Parkinson’s Disease. Biomolecules 2022; 12:biom12020243. [PMID: 35204744 PMCID: PMC8961531 DOI: 10.3390/biom12020243] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 12/02/2022] Open
Abstract
The discovery of the D3 receptor (D3R) subtypes of dopamine (DA) has generated an understandable increase in interest in the field of neurological diseases, especially Parkinson’s disease (PD). Indeed, although DA replacement therapy with l-DOPA has provided an effective treatment for patients with PD, it is responsible for invalidating abnormal involuntary movements, known as L-DOPA-induced dyskinesia, which constitutes a serious limitation of the use of this therapy. Of particular interest is the finding that chronic l-DOPA treatment can trigger the expression of D1R–D3R heteromeric interactions in the dorsal striatum. The D3R is expressed in various tissues of the central nervous system, including the striatum. Compelling research has focused on striatal D3Rs in the context of PD and motor side effects, including dyskinesia, occurring with DA replacement therapy. Therefore, this review will briefly describe the basal ganglia (BG) and the DA transmission within these brain regions, before going into more detail with regard to the role of D3Rs in PD and their participation in the current treatments. Numerous studies have also highlighted specific interactions between D1Rs and D3Rs that could promote dyskinesia. Finally, this review will also address the possibility that D3Rs located outside of the BG may mediate some of the effects of DA replacement therapy.
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Différenciation et Communication Neuroendocrine, Endocrine et Germinale Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), University of Rouen, INSERM 1239, 76000 Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, 76000 Rouen, France
- Correspondence: ; Tel.: +33-2-35-14-83-69
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2080 Msida, Malta;
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Philippe De Deurwaerdère
- Unité Mixte de Recherche (UMR) 5287, Centre National de la Recherche Scientifique (CNRS), CEDEX, 33000 Bordeaux, France;
| |
Collapse
|
10
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
11
|
Jones-Tabah J, Martin RD, Chen JJ, Tanny JC, Clarke PBS, Hébert TE. A role for BET proteins in regulating basal, dopamine-induced and cAMP/PKA-dependent transcription in rat striatal neurons. Cell Signal 2021; 91:110226. [PMID: 34974082 DOI: 10.1016/j.cellsig.2021.110226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
The activity of striatal medium-spiny projection neurons is regulated by D1 and D2 dopamine receptors. The D1 receptor (D1R) is a Gαs/olf-coupled GPCR which activates a cAMP/PKA/DARPP-32 signalling cascade that increases excitability and facilitates plasticity, partly through the regulation of transcription. Upon activation via D1R, PKA can translocate to the nucleus to regulate transcription through the phosphorylation of various targets. One candidate effector of PKA-dependent transcriptional regulation is the BET protein Brd4. It is known that when Brd4 is activated by phosphorylation, it binds more readily to acetylated histones at promoters and enhancers; moreover, in non-neuronal cells, PKA signalling has been shown to increase recruitment of Brd4 to chromatin. However, it is unknown whether BET proteins, or Brd4 specifically, are involved in transcriptional activation by cAMP/PKA in neurons. Here, we demonstrate that in adult rats, inhibition of BET proteins with the bromodomain inhibitor JQ1 suppressed the expression of ~25% of D1R-upregulated genes, while also increasing the expression of a subset of immediate-early genes. We further found that cAMP/PKA signalling promotes Brd4 recruitment to dopamine-induced genes in striatal neurons, and that knockdown of Brd4 attenuates D1R-induced gene expression. Finally, we report that JQ1 treatment downregulated expression of many GPCRs and also impaired ERK1/2 signalling in striatal neurons. Our findings identify the BET protein family, and Brd4 in particular, as novel regulators of basal and D1R-dependent transcription in rat striatal neurons, and delineate complex bi-directional effects of bromodomain inhibitors on neuronal transcription.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jennifer J Chen
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
12
|
Altwal F, Padovan-Neto FE, Ritger A, Steiner H, West AR. Role of 5-HT1A Receptor in Vilazodone-Mediated Suppression of L-DOPA-Induced Dyskinesia and Increased Responsiveness to Cortical Input in Striatal Medium Spiny Neurons in an Animal Model of Parkinson's Disease. Molecules 2021; 26:molecules26195790. [PMID: 34641332 PMCID: PMC8510243 DOI: 10.3390/molecules26195790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/20/2021] [Indexed: 01/06/2023] Open
Abstract
L-DOPA therapy in Parkinson’s disease (PD) is limited due to emerging L-DOPA-induced dyskinesia. Research has identified abnormal dopamine release from serotonergic (5-HT) terminals contributing to this dyskinesia. Selective serotonin reuptake inhibitors (SSRIs) or 5-HT receptor (5-HTr) agonists can regulate 5-HT activity and attenuate dyskinesia, but they often also produce a loss of the antiparkinsonian efficacy of L-DOPA. We investigated vilazodone, a novel multimodal 5-HT agent with SSRI and 5-HTr1A partial agonist properties, for its potential to reduce dyskinesia without interfering with the prokinetic effects of L-DOPA, and underlying mechanisms. We assessed vilazodone effects on L-DOPA-induced dyskinesia (abnormal involuntary movements, AIMs) and aberrant responsiveness to corticostriatal drive in striatal medium spiny neurons (MSNs) measured with in vivo single-unit extracellular recordings, in the 6-OHDA rat model of PD. Vilazodone (10 mg/kg) suppressed all subtypes (axial, limb, orolingual) of AIMs induced by L-DOPA (5 mg/kg) and the increase in MSN responsiveness to cortical stimulation (shorter spike onset latency). Both the antidyskinetic effects and reversal in MSN excitability by vilazodone were inhibited by the 5-HTr1A antagonist WAY-100635, demonstrating a critical role for 5-HTr1A in these vilazodone actions. Our results indicate that vilazodone may serve as an adjunct therapeutic for reducing dyskinesia in patients with PD.
Collapse
Affiliation(s)
- Feras Altwal
- Center for Neurodegenerative Disease & Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (F.A.); (A.R.W.)
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
| | - Fernando E. Padovan-Neto
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
| | - Alexandra Ritger
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Correspondence:
| | - Anthony R. West
- Center for Neurodegenerative Disease & Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (F.A.); (A.R.W.)
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
| |
Collapse
|
13
|
Scheggi S, Rossi F, Corsi S, Fanni S, Tronci E, Ludovica C, Vargiu R, Gambarana C, Muñoz A, Stancampiano R, Björklund A, Carta M. BDNF Overexpression Increases Striatal D3 Receptor Level at Striatal Neurons and Exacerbates D1-Receptor Agonist-Induced Dyskinesia. JOURNAL OF PARKINSONS DISEASE 2021; 10:1503-1514. [PMID: 32651332 DOI: 10.3233/jpd-202061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND We recently showed that striatal overexpression of brain derived neurotrophic factor (BDNF) by adeno-associated viral (AAV) vector exacerbated L-DOPA-induced dyskinesia (LID) in 6-OHDA-lesioned rats. An extensive sprouting of striatal serotonergic terminals accompanied this effect, accounting for the increased susceptibility to LID. OBJECTIVE We set to investigate whether the BDNF effect was restricted to LID, or extended to dyskinesia induced by direct D1 receptor agonists. METHODS Unilaterally 6-OHDA-lesioned rats received a striatal injection of an AAV vector to induce BDNF or GFP overexpression. Eight weeks later, animals received daily treatments with a low dose of SKF82958 (0.02 mg/kg s.c.) and development of dyskinesia was evaluated. At the end of the experiment, D1 and D3 receptors expression levels and D1 receptor-dependent signaling pathways were measured in the striatum. RESULTS BDNF overexpression induced significant worsening of dyskinesia induced by SKF82958 compared to the GFP group and increased the expression of D3 receptor at striatal level, even in absence of pharmacological treatment; by contrast, D1 receptor levels were not affected. In BDNF-overexpressing striata, SKF82958 administration resulted in increased levels of D1-D3 receptors co-immunoprecipitation and increased phosphorylation levels of Thr34 DARPP-32 and ERK1/2. CONCLUSION Here we provide evidence for a functional link between BDNF, D3 receptors and D1-D3 receptor close interaction in the augmented susceptibility to dyskinesia in 6-OHDA-lesioned rats. We suggest that D1-D3 receptors interaction may be instrumental in driving the molecular alterations underlying the appearance of dyskinesia; its disruption may be a therapeutic strategy for treating dyskinesia in PD patients.
Collapse
Affiliation(s)
- Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Francesca Rossi
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Sara Corsi
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Congiu Ludovica
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Romina Vargiu
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ana Muñoz
- Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Anders Björklund
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology, Lund University, Lund, Sweden
| | - Manolo Carta
- Department of Biomedical Sciences, Cagliari University, Cagliari, Italy
| |
Collapse
|
14
|
Urakubo H, Yagishita S, Kasai H, Kubota Y, Ishii S. The critical balance between dopamine D2 receptor and RGS for the sensitive detection of a transient decay in dopamine signal. PLoS Comput Biol 2021; 17:e1009364. [PMID: 34591840 PMCID: PMC8483376 DOI: 10.1371/journal.pcbi.1009364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/18/2021] [Indexed: 12/19/2022] Open
Abstract
In behavioral learning, reward-related events are encoded into phasic dopamine (DA) signals in the brain. In particular, unexpected reward omission leads to a phasic decrease in DA (DA dip) in the striatum, which triggers long-term potentiation (LTP) in DA D2 receptor (D2R)-expressing spiny-projection neurons (D2 SPNs). While this LTP is required for reward discrimination, it is unclear how such a short DA-dip signal (0.5-2 s) is transferred through intracellular signaling to the coincidence detector, adenylate cyclase (AC). In the present study, we built a computational model of D2 signaling to determine conditions for the DA-dip detection. The DA dip can be detected only if the basal DA signal sufficiently inhibits AC, and the DA-dip signal sufficiently disinhibits AC. We found that those two requirements were simultaneously satisfied only if two key molecules, D2R and regulators of G protein signaling (RGS) were balanced within a certain range; this balance has indeed been observed in experimental studies. We also found that high level of RGS was required for the detection of a 0.5-s short DA dip, and the analytical solutions for these requirements confirmed their universality. The imbalance between D2R and RGS is associated with schizophrenia and DYT1 dystonia, both of which are accompanied by abnormal striatal LTP. Our simulations suggest that D2 SPNs in patients with schizophrenia and DYT1 dystonia cannot detect short DA dips. We finally discussed that such psychiatric and movement disorders can be understood in terms of the imbalance between D2R and RGS.
Collapse
Affiliation(s)
- Hidetoshi Urakubo
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
- Section of Electron Microscopy, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan
| | - Yoshiyuki Kubota
- Section of Electron Microscopy, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| | - Shin Ishii
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan
| |
Collapse
|
15
|
Mechanisms of Antiparkinsonian Anticholinergic Therapy Revisited. Neuroscience 2021; 467:201-217. [PMID: 34048797 DOI: 10.1016/j.neuroscience.2021.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 01/15/2023]
Abstract
Before the advent of L-DOPA, the gold standard symptomatic therapy for Parkinson's disease (PD), anticholinergic drugs (muscarinic receptor antagonists) were the preferred antiparkinsonian therapy, but their unwanted side effects associated with impaired extrastriatal cholinergic function limited their clinical utility. Since most patients treated with L-DOPA also develop unwanted side effects such as L-DOPA-induced dyskinesia (LID), better therapies are needed. Recent studies in animal models demonstrate that optogenetic and chemogenetic manipulation of striatal cholinergic interneurons (SCIN), the main source of striatal acetylcholine, modulate parkinsonism and LID, suggesting that restoring SCIN function might serve as a therapeutic option that avoids extrastriatal anticholinergics' side effects. However, it is still unclear how the altered SCIN activity in PD and LID affects the striatal circuit, whereas the mechanisms of action of anticholinergic drugs are still not fully understood. Recent animal model studies showing that SCINs undergo profound changes in their tonic discharge pattern after chronic L-DOPA administration call for a reexamination of classical views of how SCINs contribute to PD symptoms and LID. Here, we review the recent advances on the circuit implications of aberrant striatal cholinergic signaling in PD and LID in an effort to provide a comprehensive framework to understand the effects of anticholinergic drugs and with the aim of shedding light into future perspectives of cholinergic circuit-based therapies.
Collapse
|
16
|
Correlación entre la expresión del receptor dopaminérgico D2 y presencia de movimientos involuntarios anormales (MIA) en un modelo de disquinesia en ratas Wistar hemiparkinsonizadas. Neurologia 2021. [DOI: 10.1016/j.nrl.2017.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
17
|
Caro Aponte P, Otálora C, Guzmán J, Turner L, Alcázar J, Mayorga E. Correlation between dopamine receptor D2 expression and presence of abnormal involuntary movements in Wistar rats with hemiparkinsonism and dyskinesia. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2017.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
18
|
Paz RM, Tubert C, Stahl AM, Amarillo Y, Rela L, Murer MG. Levodopa Causes Striatal Cholinergic Interneuron Burst-Pause Activity in Parkinsonian Mice. Mov Disord 2021; 36:1578-1591. [PMID: 33547844 DOI: 10.1002/mds.28516] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Enhanced striatal cholinergic interneuron activity contributes to the striatal hypercholinergic state in Parkinson's disease (PD) and to levodopa-induced dyskinesia. In severe PD, dyskinesia and motor fluctuations become seriously debilitating, and the therapeutic strategies become scarce. Given that the systemic administration of anticholinergics can exacerbate extrastriatal-related symptoms, targeting cholinergic interneurons is a promising therapeutic alternative. Therefore, unraveling the mechanisms causing pathological cholinergic interneuron activity in severe PD with motor fluctuations and dyskinesia may provide new molecular therapeutic targets. METHODS We used ex vivo electrophysiological recordings combined with pharmacological and morphological studies to investigate the intrinsic alterations of cholinergic interneurons in the 6-hydroxydopamine mouse model of PD treated with levodopa. RESULTS Cholinergic interneurons exhibit pathological burst-pause activity in the parkinsonian "off levodopa" state. This is mediated by a persistent ligand-independent activity of dopamine D1/D5 receptor signaling, involving a cyclic adenosine monophosphate (cAMP) pathway. Dysregulation of membrane ion channels that results in increased inward-rectifier potassium type 2 (Kir2) and decreased leak currents causes the burst pause activity, which can be dampened by pharmacological inhibition of intracellular cAMP. A single challenge with a dyskinetogenic dose of levodopa is sufficient to induce persistent cholinergic interneuron burst-pause firing. CONCLUSION Our data unravel a mechanism causing aberrant cholinergic interneuron burst-pause activity in parkinsonian mice treated with levodopa. Targeting D5-cAMP signaling and the regulation of Kir2 and leak channels may alleviate parkinsonism and dyskinesia by restoring normal cholinergic interneuron function. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rodrigo Manuel Paz
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, 2155 Paraguay Street, Buenos Aires, 1121, Argentina
| | - Cecilia Tubert
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, 2155 Paraguay Street, Buenos Aires, 1121, Argentina
| | - Agostina Monica Stahl
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, 2155 Paraguay Street, Buenos Aires, 1121, Argentina
| | - Yimy Amarillo
- Departamento de Física Médica, Centro Atómico Bariloche and Instituto Balseiro, CONICET, 9500 Ezequiel Bustillo Avenue, San Carlos de Bariloche, Rio Negro, 8402, Argentina
| | - Lorena Rela
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, 2155 Paraguay Street, Buenos Aires, 1121, Argentina
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, 2155 Paraguay Street, Buenos Aires, 1121, Argentina
| |
Collapse
|
19
|
Sebastianutto I, Goyet E, Andreoli L, Font-Ingles J, Moreno-Delgado D, Bouquier N, Jahannault-Talignani C, Moutin E, Di Menna L, Maslava N, Pin JP, Fagni L, Nicoletti F, Ango F, Cenci MA, Perroy J. D1-mGlu5 heteromers mediate noncanonical dopamine signaling in Parkinson's disease. J Clin Invest 2020; 130:1168-1184. [PMID: 32039920 DOI: 10.1172/jci126361] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Dopamine receptor D1 modulates glutamatergic transmission in cortico-basal ganglia circuits and represents a major target of L-DOPA therapy in Parkinson's disease. Here we show that D1 and metabotropic glutamate type 5 (mGlu5) receptors can form previously unknown heteromeric entities with distinctive functional properties. Interacting with Gq proteins, cell-surface D1-mGlu5 heteromers exacerbated PLC signaling and intracellular calcium release in response to either glutamate or dopamine. In rodent models of Parkinson's disease, D1-mGlu5 nanocomplexes were strongly upregulated in the dopamine-denervated striatum, resulting in a synergistic activation of PLC signaling by D1 and mGlu5 receptor agonists. In turn, D1-mGlu5-dependent PLC signaling was causally linked with excessive activation of extracellular signal-regulated kinases in striatal neurons, leading to dyskinesia in animals treated with L-DOPA or D1 receptor agonists. The discovery of D1-mGlu5 functional heteromers mediating maladaptive molecular and motor responses in the dopamine-denervated striatum may prompt the development of new therapeutic principles for Parkinson's disease.
Collapse
Affiliation(s)
- Irene Sebastianutto
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elise Goyet
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Laura Andreoli
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joan Font-Ingles
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - David Moreno-Delgado
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Department of Neuroscience Research, UCB Pharma, Braine l'Alleud, Belgium
| | - Nathalie Bouquier
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Enora Moutin
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Luisa Di Menna
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Natallia Maslava
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Laurent Fagni
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Ferdinando Nicoletti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Fabrice Ango
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Julie Perroy
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
20
|
The Multimodal Serotonergic Agent Vilazodone Inhibits L-DOPA-Induced Gene Regulation in Striatal Projection Neurons and Associated Dyskinesia in an Animal Model of Parkinson's Disease. Cells 2020; 9:cells9102265. [PMID: 33050305 PMCID: PMC7600385 DOI: 10.3390/cells9102265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
Levodopa (L-DOPA) treatment in Parkinson's disease is limited by the emergence of L-DOPA-induced dyskinesia. Such dyskinesia is associated with aberrant gene regulation in neurons of the striatum, which is caused by abnormal dopamine release from serotonin terminals. Previous work showed that modulating the striatal serotonin innervation with selective serotonin reuptake inhibitors (SSRIs) or 5-HT1A receptor agonists could attenuate L-DOPA-induced dyskinesia. We investigated the effects of a novel serotonergic agent, vilazodone, which combines SSRI and 5-HT1A partial agonist properties, on L-DOPA-induced behavior and gene regulation in the striatum in an animal model of Parkinson's disease. After unilateral dopamine depletion by 6-hydroxydopamine (6-OHDA), rats received repeated L-DOPA treatment (5 mg/kg) alone or in combination with vilazodone (10 mg/kg) for 3 weeks. Gene regulation was then mapped throughout the striatum using in situ hybridization histochemistry. Vilazodone suppressed the development of L-DOPA-induced dyskinesia and turning behavior but did not interfere with the prokinetic effects of L-DOPA (forelimb stepping). L-DOPA treatment drastically increased the expression of dynorphin (direct pathway), 5-HT1B, and zif268 mRNA in the striatum ipsilateral to the lesion. These effects were inhibited by vilazodone. In contrast, vilazodone had no effect on enkephalin expression (indirect pathway) or on gene expression in the intact striatum. Thus, vilazodone inhibited L-DOPA-induced gene regulation selectively in the direct pathway of the dopamine-depleted striatum, molecular changes that are considered critical for L-DOPA-induced dyskinesia. These findings position vilazodone, an approved antidepressant, as a potential adjunct medication for the treatment of L-DOPA-induced motor side effects.
Collapse
|
21
|
Nakamura Y, Nakamura Y, Pelosi A, Djemai B, Debacker C, Hervé D, Girault JA, Tsurugizawa T. fMRI detects bilateral brain network activation following unilateral chemogenetic activation of direct striatal projection neurons. Neuroimage 2020; 220:117079. [DOI: 10.1016/j.neuroimage.2020.117079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/23/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
|
22
|
Kanzato N, Nakachi K, Naka T, Mochizuki S, Miyamae Y, Okada Y. Parkinson's disease therapy with Istradefylline and blood biomarkers of epigenetics. ACTA ACUST UNITED AC 2020. [DOI: 10.1111/ncn3.12415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Naomi Kanzato
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Kou Nakachi
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Taro Naka
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Satsuki Mochizuki
- Department of Surgery National Defense Medical College Saitama Japan
| | - Yuka Miyamae
- Department of Pathophysiology for Locomotive and Neoplastic Diseases Juntendo University Graduate School of Medicine Tokyo Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases Juntendo University Graduate School of Medicine Tokyo Japan
| |
Collapse
|
23
|
Padovan-Neto FE, Patterson S, F Voelkner NM, Altwal F, Beverley JA, West AR, Steiner H. Selective Regulation of 5-HT1B Serotonin Receptor Expression in the Striatum by Dopamine Depletion and Repeated L-DOPA Treatment: Relationship to L-DOPA-Induced Dyskinesias. Mol Neurobiol 2020; 57:736-751. [PMID: 31468338 PMCID: PMC7035192 DOI: 10.1007/s12035-019-01739-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/16/2019] [Indexed: 11/28/2022]
Abstract
Dopamine and serotonin in the basal ganglia interact in a bidirectional manner. On the one hand, serotonin (5-HT) receptors regulate the effects of dopamine agonists on several levels, ranging from molecular signaling to behavior. These interactions include 5-HT receptor-mediated facilitation of dopamine receptor-induced gene regulation in striatal output pathways, which involves the 5-HT1B receptor and others. Conversely, there is evidence that dopamine action by psychostimulants regulates 5-HT1B receptor expression in the striatum. To further investigate the effects of dopamine and agonists on 5-HT receptors, we assessed the expression of 5-HT1B and other serotonin receptor subtypes in the striatum after unilateral dopamine depletion by 6-OHDA and subsequent treatment with L-DOPA (5 mg/kg; 4 weeks). Neither dopamine depletion nor L-DOPA treatment produced significant changes in 5-HT2C, 5-HT4, or 5-HT6 receptor expression in the striatum. In contrast, the 6-OHDA lesion caused a (modest) increase in 5-HT1B mRNA levels throughout the striatum. Moreover, repeated L-DOPA treatment markedly further elevated 5-HT1B expression in the dopamine-depleted striatum, an effect that was most robust in the sensorimotor striatum. A minor L-DOPA-induced increase in 5-HT1B expression was also seen in the intact striatum. These changes in 5-HT1B expression mimicked changes in the expression of neuropeptide markers (dynorphin, enkephalin mRNA) in striatal projection neurons. After repeated L-DOPA treatment, the severity of L-DOPA-induced dyskinesias and turning behavior was positively correlated with the increase in 5-HT1B expression in the associative, but not sensorimotor, striatum ipsilateral to the lesion, suggesting that associative striatal 5-HT1B receptors may play a role in L-DOPA-induced behavioral abnormalities.
Collapse
Affiliation(s)
- Fernando E Padovan-Neto
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, 14040-901, SP, Brazil
| | - Santanna Patterson
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Nivea M F Voelkner
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Feras Altwal
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Joel A Beverley
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Anthony R West
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Heinz Steiner
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
24
|
Noninvasive Brain Stimulation Enhances Memory Acquisition and Is Associated with Synaptoneurosome Modification in the Rat Hippocampus. eNeuro 2019; 6:ENEURO.0311-19.2019. [PMID: 31699891 PMCID: PMC6900464 DOI: 10.1523/eneuro.0311-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 10/31/2019] [Indexed: 01/11/2023] Open
Abstract
Transcranial direct-current stimulation (tDCS) is a non-invasive brain stimulation approach previously shown to enhance memory acquisition, but more studies are needed to elucidate the underlying mechanisms. Here, we examined the effects of anodal tDCS (0.25 mA for 30 min) on the memory performance of male Sprague Dawley rats in the passive avoidance test (PAT) and the associated modifications to the hippocampal proteomes. Results indicate anodal tDCS applied before the acquisition period significantly enhanced memory performance in the PAT. Following PAT, synaptoneurosomes were biochemically purified from the hippocampi of tDCS-treated or sham-treated rats and individual protein abundances were determined by bottom-up liquid chromatography mass spectrometry analysis. Proteomic analysis identified 184 differentially expressed hippocampal proteins when comparing the sham to the tDCS before memory acquisition treatment group. Ingenuity pathway analysis (IPA) showed anodal tDCS before memory acquisition significantly enhanced pathways associated with memory, cognition, learning, transmission, neuritogenesis, and long-term potentiation (LTP). IPA identified significant upstream regulators including bdnf, shank3, and gsk3b. Protein-protein interaction (PPI) and protein sequence similarity (PSS) networks show that glutamate receptor pathways, ion channel activity, memory, learning, cognition, and long-term memory were significantly associated with anodal tDCS. Centrality measures from both networks identified key proteins including dlg, shank, grin, and gria that were significantly modified by tDCS applied before the acquisition period. Together, our results provide descriptive molecular evidence that anodal tDCS enhances memory performance in the PAT by modifying hippocampal synaptic plasticity related proteins.
Collapse
|
25
|
Keifman E, Ruiz-DeDiego I, Pafundo DE, Paz RM, Solís O, Murer MG, Moratalla R. Optostimulation of striatonigral terminals in substantia nigra induces dyskinesia that increases after L-DOPA in a mouse model of Parkinson's disease. Br J Pharmacol 2019; 176:2146-2161. [PMID: 30895594 DOI: 10.1111/bph.14663] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/08/2019] [Accepted: 02/20/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND AND PURPOSE L-DOPA-induced dyskinesia (LID) remains a major complication of L-DOPA therapy in Parkinson's disease. LID is believed to result from inhibition of substantia nigra reticulata (SNr) neurons by GABAergic striatal projection neurons that become supersensitive to dopamine receptor stimulation after severe nigrostriatal degeneration. Here, we asked if stimulation of direct medium spiny neuron (dMSN) GABAergic terminals at the SNr can produce a full dyskinetic state similar to that induced by L-DOPA. EXPERIMENTAL APPROACH Adult C57BL6 mice were lesioned with 6-hydroxydopamine in the medial forebrain bundle. Channel rhodopsin was expressed in striatonigral terminals by ipsilateral striatal injection of adeno-associated viral particles under the CaMKII promoter. Optic fibres were implanted on the ipsilateral SNr. Optical stimulation was performed before and 24 hr after three daily doses of L-DOPA at subthreshold and suprathreshold dyskinetic doses. We also examined the combined effect of light stimulation and an acute L-DOPA challenge. KEY RESULTS Optostimulation of striatonigral terminals inhibited SNr neurons and induced all dyskinesia subtypes (optostimulation-induced dyskinesia [OID]) in 6-hydroxydopamine animals, but not in sham-lesioned animals. Additionally, chronic L-DOPA administration sensitised dyskinetic responses to striatonigral terminal optostimulation, as OIDs were more severe 24 hr after L-DOPA administration. Furthermore, L-DOPA combined with light stimulation did not result in higher dyskinesia scores than OID alone, suggesting that optostimulation has a masking effect on LID. CONCLUSION AND IMPLICATIONS This work suggests that striatonigral inhibition of basal ganglia output (SNr) is a decisive mechanism mediating LID and identifies the SNr as a target for managing LID.
Collapse
Affiliation(s)
- Ettel Keifman
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Irene Ruiz-DeDiego
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Diego Esteban Pafundo
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Rodrigo Manuel Paz
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| |
Collapse
|
26
|
Genetic enhancement of Ras-ERK pathway does not aggravate L-DOPA-induced dyskinesia in mice but prevents the decrease induced by lovastatin. Sci Rep 2018; 8:15381. [PMID: 30337665 PMCID: PMC6194127 DOI: 10.1038/s41598-018-33713-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/03/2018] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence supports a close relationship between Ras-ERK1/2 activation in the striatum and L-DOPA-induced dyskinesia (LID). ERK1/2 activation by L-DOPA takes place through the crosstalk between D1R/AC/PKA/DARPP-32 pathway and NMDA/Ras pathway. Compelling genetic and pharmacological evidence indicates that Ras-ERK1/2 inhibition prevents LID onset and may even revert already established dyskinetic symptoms. However, it is currently unclear whether exacerbation of Ras-ERK1/2 activity in the striatum may further aggravate dyskinesia in experimental animal models. Here we took advantage of two genetic models in which Ras-ERK1/2 signaling is hyperactivated, the Nf1+/− mice, in which the Ras inhibitor neurofibromin is reduced, and the Ras-GRF1 overexpressing (Ras-GRF1 OE) transgenic mice in which a specific neuronal activator of Ras is enhanced. Nf1+/− and Ras-GRF1 OE mice were unilaterally lesioned with 6-OHDA and treated with an escalating L-DOPA dosing regimen. In addition, a subset of Nf1+/− hemi-parkinsonian animals was also co-treated with the Ras inhibitor lovastatin. Our results revealed that Nf1+/− and Ras-GRF1 OE mice displayed similar dyskinetic symptoms to their wild-type counterparts. This observation was confirmed by the lack of differences between mutant and wild-type mice in striatal molecular changes associated to LID (i.e., FosB, and pERK1/2 expression). Interestingly, attenuation of Ras activity with lovastatin does not weaken dyskinetic symptoms in Nf1+/− mice. Altogether, these data suggest that ERK1/2-signaling activation in dyskinetic animals is maximal and does not require further genetic enhancement in the upstream Ras pathway. However, our data also demonstrate that such a genetic enhancement may reduce the efficacy of anti-dyskinetic drugs like lovastatin.
Collapse
|
27
|
Huang M, Bai M, Zhang Z, Ge L, Lu K, Li X, Li Y, Zhou X, Guo N, Yang L, Bai J. Downregulation of thioredoxin-1 in the ventral tegmental area delays extinction of methamphetamine-induced conditioned place preference. J Psychopharmacol 2018; 32:1037-1046. [PMID: 30136629 DOI: 10.1177/0269881118791523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Drug addiction is characterized by compulsive drug use and relapse. Thioredoxin-1 is emerging as an important modulator involved in the cellular protective response against a variety of toxic stressors. Previous study has reported that thioredoxin-1 overexpression prevents the acquisition of methamphetamine-conditioned place preference. Here, we aimed to investigate the effect of thioredoxin-1 on methamphetamine-conditioned place preference extinction and the possible mechanism. METHODS (a) An extinction procedure in mice was employed to investigate the effect of thioredoxin-1 on the extinction of methamphetamine-conditioned place preference. After the acquisition of methamphetamine-conditioned place preference, mice underwent the following procedures: the injection of thioredoxin-1 small interfering RNA in the ventral tegmental area followed by the post-conditioned place preference test, four days of extinction training followed by four days of recovery after surgery. (b) The levels of thioredoxin-1, dopamine D1 receptor, tyrosine hydroxylase, phosphorylated extracellular regulated kinase, and phosphorylated cyclic adenosine monophosphate response element binding protein were examined by using Western blot analysis. RESULTS Thioredoxin-1 downregulation in the ventral tegmental area delayed methamphetamine-conditioned place preference extinction. The expression of thioredoxin-1 was decreased in the ventral tegmental area of mice in control and negative groups after methamphetamine-conditioned place preference extinction, but not in the thioredoxin-1 siRNA group. The levels of dopamine D1 receptor, tyrosine hydroxylase, phosphorylated extracellular regulated kinase, and phosphorylated cyclic adenosine monophosphate response element binding protein were decreased in the ventral tegmental area, nucleus accumbens, and prefrontal cortex of mice in the control and negative groups after methamphetamine-conditioned place preference extinction, but were inversely increased in thioredoxin-1 siRNA group. CONCLUSIONS The results suggest that downregulation of thioredoxin-1 in the ventral tegmental area may delay methamphetamine-conditioned place preference extinction by regulating the mesocorticolimbic dopaminergic signaling pathway.
Collapse
Affiliation(s)
- Mengbing Huang
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China.,2 Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Ming Bai
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Zhimin Zhang
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Lu Ge
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Kang Lu
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Xiang Li
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China.,2 Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Ye Li
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Xiaoshuang Zhou
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Ningning Guo
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China.,2 Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Lihua Yang
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China.,3 Narcotics Control School, Yunnan Police College, Kunming, China
| | - Jie Bai
- 1 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
28
|
Bordia T, Perez XA. Cholinergic control of striatal neurons to modulate L-dopa-induced dyskinesias. Eur J Neurosci 2018; 49:859-868. [PMID: 29923650 DOI: 10.1111/ejn.14048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/28/2022]
Abstract
L-dopa induced dyskinesias (LIDs) are a disabling motor complication of L-dopa therapy for Parkinson's disease (PD) management. Treatment options remain limited and the underlying network mechanisms remain unclear due to a complex pathophysiology. What is well-known, however, is that aberrant striatal signaling plays a key role in LIDs development. Here, we discuss the specific contribution of striatal cholinergic interneurons (ChIs) and GABAergic medium spiny projection neurons (MSNs) with a particular focus on how cholinergic signaling may integrate multiple striatal systems to modulate LIDs expression. Enhanced ChI transmission, altered MSN activity and the associated abnormal downstream signaling responses that arise with nigrostriatal damage are well known to contribute to LIDs development. In fact, enhancing M4 muscarinic receptor activity, a receptor favorably expressed on D1 dopamine receptor-expressing MSNs dampens their activity to attenuate LIDs. Likewise, ChI activation via thalamostriatal neurons is shown to interrupt cortical signaling to enhance D2 dopamine receptor-expressing MSN activity via M1 muscarinic receptors, which may interrupt ongoing motor activity. Notably, numerous preclinical studies also show that reducing nicotinic cholinergic receptor activity decreases LIDs. Taken together, these studies indicate the importance of cholinergic control of striatal neuronal activity and point to muscarinic and nicotinic receptors as significant pharmacological targets for alleviating LIDs in PD patients.
Collapse
Affiliation(s)
- Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025, USA
| | - Xiomara A Perez
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025, USA
| |
Collapse
|
29
|
Differential Synaptic Remodeling by Dopamine in Direct and Indirect Striatal Projection Neurons in Pitx3 -/- Mice, a Genetic Model of Parkinson's Disease. J Neurosci 2018; 38:3619-3630. [PMID: 29483281 DOI: 10.1523/jneurosci.3184-17.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/23/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
In toxin-based models of Parkinson's disease (PD), striatal projection neurons (SPNs) exhibit dendritic atrophy and spine loss concurrent with an increase in excitability. Chronic l-DOPA treatment that induces dyskinesia selectively restores spine density and excitability in indirect pathway SPNs (iSPNs), whereas spine loss and hyperexcitability persist in direct pathway SPNs (dSPNs). These alterations have only been characterized in toxin-based models of PD, raising the possibility that they are an artifact of exposure to the toxin, which may engage compensatory mechanisms independent of the PD-like pathology or due to the loss of dopaminergic afferents. To test all these, we studied the synaptic remodeling in Pitx3-/- or aphakia mice, a genetic model of PD, in which most of the dopamine neurons in the substantia nigra fail to fully differentiate and to innervate the striatum. We made 3D reconstructions of the dendritic arbor and measured excitability in identified SPNs located in dorsal striatum of BAC-Pitx3-/- mice treated with saline or l-DOPA. Both dSPNs and iSPNs from BAC-Pitx3-/- mice had shorter dendritic trees, lower spine density, and more action potentials than their counterparts from WT mice. Chronic l-DOPA treatment restored spine density and firing rate in iSPNs. By contrast, in dSPNs, spine loss and hyperexcitability persisted following l-DOPA treatment, which is similar to what happens in 6-OHDA WT mice. This indicates that dopamine-mediated synaptic remodeling and plasticity is independent of dopamine innervation during SPN development and that Pitx3-/- mice are a good model because they develop the same pathology described in the toxins-based models and in human postmortem studies of advanced PD.SIGNIFICANCE STATEMENT As the only genetic model of Parkinson's disease (PD) that develops dyskinesia, Pitx3-/- mice reproduce the behavioral effects seen in humans and are a good system for studying dopamine-induced synaptic remodeling. The studies we present here establish that the structural and functional synaptic plasticity that occur in striatal projection neurons in PD and in l-DOPA-induced dyskinesia are specifically due to modulation of the neurotransmitter dopamine and are not artifacts of the use of chemical toxins in PD models. In addition, our findings provide evidence that synaptic plasticity in the Pitx3-/- mouse is similar to that seen in toxin models despite its lack of dopaminergic innervation of the striatum during development. Pitx3-/- mice reproduced the alterations described in patients with advanced PD and in well accepted toxin-based models of PD and dyskinesia. These results further consolidate the fidelity of the Pitx3-/- mouse as a PD model in which to study the morphological and physiological remodeling of striatal projection neurons by administration of l-DOPA and other drugs.
Collapse
|
30
|
Solís O, Moratalla R. Dopamine receptors: homomeric and heteromeric complexes in l-DOPA-induced dyskinesia. J Neural Transm (Vienna) 2018; 125:1187-1194. [DOI: 10.1007/s00702-018-1852-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
|
31
|
Gomez G, Saborido MD, Bernardi MA, Gershanik OS, Taravini IR, Ferrario JE. Regulation of Pleiotrophin and Fyn in the striatum of rats undergoing L-DOPA-induced dyskinesia. Neurosci Lett 2018; 666:5-10. [DOI: 10.1016/j.neulet.2017.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/01/2017] [Accepted: 12/10/2017] [Indexed: 12/20/2022]
|
32
|
Suarez LM, Solis O, Aguado C, Lujan R, Moratalla R. L-DOPA Oppositely Regulates Synaptic Strength and Spine Morphology in D1 and D2 Striatal Projection Neurons in Dyskinesia. Cereb Cortex 2018; 26:4253-4264. [PMID: 27613437 PMCID: PMC5066835 DOI: 10.1093/cercor/bhw263] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/01/2016] [Indexed: 12/21/2022] Open
Abstract
Dopamine depletion in Parkinson's disease (PD) produces dendritic spine loss in striatal medium spiny neurons (MSNs) and increases their excitability. However, the synaptic changes that occur in MSNs in PD, in particular those induced by chronic L-3,4-dihydroxyphenylalanine (L-DOPA) treatment, are still poorly understood. We exposed BAC-transgenic D1-tomato and D2-eGFP mice to PD and dyskinesia model paradigms, enabling cell type-specific assessment of changes in synaptic physiology and morphology. The distinct fluorescence markers allowed us to identify D1 and D2 MSNs for analysis using intracellular sharp electrode recordings, electron microscopy, and 3D reconstructions with single-cell Lucifer Yellow injections. Dopamine depletion induced spine pruning in both types of MSNs, affecting mushroom and thin spines equally. Dopamine depletion also increased firing rate in both D1- and D2-MSNs, but reduced evoked-EPSP amplitude selectively in D2-MSNs. L-DOPA treatment that produced dyskinesia differentially affected synaptic properties in D1- and D2-MSNs. In D1-MSNs, spine density remained reduced but the remaining spines were enlarged, with bigger heads and larger postsynaptic densities. These morphological changes were accompanied by facilitation of action potential firing triggered by synaptic inputs. In contrast, although L-DOPA restored the number of spines in D2-MSNs, it resulted in shortened postsynaptic densities. These changes in D2-MSNs correlated with a decrease in synaptic transmission. Our findings indicate that L-DOPA-induced dyskinesia is associated with abnormal spine morphology, modified synaptic transmission, and altered EPSP-spike coupling, with distinct effects in D1- and D2-MSNs.
Collapse
Affiliation(s)
- Luz M Suarez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Oscar Solis
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Carolina Aguado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Dept. Ciencias Medicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete, Spain, Spain
| | - Rafael Lujan
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Dept. Ciencias Medicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete, Spain, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| |
Collapse
|
33
|
Solís O, Garcia-Montes JR, González-Granillo A, Xu M, Moratalla R. Dopamine D3 Receptor Modulates l-DOPA-Induced Dyskinesia by Targeting D1 Receptor-Mediated Striatal Signaling. Cereb Cortex 2018; 27:435-446. [PMID: 26483399 DOI: 10.1093/cercor/bhv231] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The dopamine D3 receptor (D3R) belongs to the dopamine D2-like receptor family and is principally located in the ventral striatum. However, previous studies reported D3R overexpression in the dorsal striatum following l-DOPA treatment in parkinsonian animals. This fact has drawn attention in the importance of D3R in l-DOPA-induced dyskinesia (LID). Here, we used D3R knockout mice to assess the role of D3R in LID and rotational sensitization in the hemiparkinsonian model. Mice lacking D3R presented a reduction in dyskinesia without interfering with the antiparkinsonian l-DOPA effect and were accompanied by a reduction in the l-DOPA-induced rotations. Interestingly, deleting D3R attenuated important molecular markers in the D1R-neurons such as FosB, extracellular signal-regulated kinase, and histone-3 (H3)-activation. Colocalization studies in D1R-tomato and D2R-green fluorescent protein BAC-transgenic mice indicated that l-DOPA-induced D3R overexpression principally occurs in D1R-containing neurons although it is also present in the D2R-neurons. Moreover, D3R pharmacological blockade with PG01037 reduced dyskinesia and the molecular markers expressed in D1R-neurons. In addition, this antagonist further reduced dyskinetic symptoms in D1R heterozygous mice, indicating a direct interaction between D1R and D3R. Together, our results demonstrate that D3R modulates the development of dyskinesia by targeting D1R-mediated intracellular signaling and suggest that decreasing D3R activity may help to ameliorate LID.
Collapse
Affiliation(s)
- Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Ruben Garcia-Montes
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Aldo González-Granillo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
| | - Ming Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
34
|
Castrioto A, Carnicella S, Fraix V, Chabardes S, Moro E, Krack P. Reversing dopaminergic sensitization. Mov Disord 2017; 32:1679-1683. [PMID: 29150871 DOI: 10.1002/mds.27213] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/01/2017] [Accepted: 08/23/2017] [Indexed: 11/07/2022] Open
Affiliation(s)
- Anna Castrioto
- Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Centre hospitalier universitaire de Grenoble, Inserm, U1216, F-38000, Grenoble, France.,Movement Disorders Unit, Neurology Department, CHU de Grenoble, Grenoble, France
| | - Sebastien Carnicella
- Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Centre hospitalier universitaire de Grenoble, Inserm, U1216, F-38000, Grenoble, France
| | - Valérie Fraix
- Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Centre hospitalier universitaire de Grenoble, Inserm, U1216, F-38000, Grenoble, France.,Movement Disorders Unit, Neurology Department, CHU de Grenoble, Grenoble, France
| | - Stéphan Chabardes
- Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Centre hospitalier universitaire de Grenoble, Inserm, U1216, F-38000, Grenoble, France.,Neurosurgery Unit, Centre hospitalier universitaire de Grenoble, Grenoble, France
| | - Elena Moro
- Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Centre hospitalier universitaire de Grenoble, Inserm, U1216, F-38000, Grenoble, France.,Movement Disorders Unit, Neurology Department, CHU de Grenoble, Grenoble, France
| | - Paul Krack
- Department of Clinical Neurosciences (Neurology), University Hospital of Geneva, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
35
|
Nishijima H, Ueno T, Funamizu Y, Ueno S, Tomiyama M. Levodopa treatment and dendritic spine pathology. Mov Disord 2017; 33:877-888. [PMID: 28880414 PMCID: PMC6667906 DOI: 10.1002/mds.27172] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with the progressive loss of nigrostriatal dopaminergic neurons. Levodopa is the most effective treatment for the motor symptoms of PD. However, chronic oral levodopa treatment can lead to various motor and nonmotor complications because of nonphysiological pulsatile dopaminergic stimulation in the brain. Examinations of autopsy cases with PD have revealed a decreased number of dendritic spines of striatal neurons. Animal models of PD have revealed altered density and morphology of dendritic spines of neurons in various brain regions after dopaminergic denervation or dopaminergic denervation plus levodopa treatment, indicating altered synaptic transmission. Recent studies using rodent models have reported dendritic spine head enlargement in the caudate‐putamen, nucleus accumbens, primary motor cortex, and prefrontal cortex in cases where chronic levodopa treatment following dopaminergic denervation induced dyskinesia‐like abnormal involuntary movement. Hypertrophy of spines results from insertion of alpha‐amino‐2,3‐dihydro‐5‐methyl‐3‐oxo‐4‐isoxazolepropanoic acid receptors into the postsynaptic membrane. Such spine enlargement indicates hypersensitivity of the synapse to excitatory inputs and is compatible with a lack of depotentiation, which is an electrophysiological hallmark of levodopa‐induced dyskinesia found in the corticostriatal synapses of dyskinetic animals and the motor cortex of dyskinetic PD patients. This synaptic plasticity may be one of the mechanisms underlying the priming of levodopa‐induced complications such as levodopa‐induced dyskinesia and dopamine dysregulation syndrome. Drugs that could potentially prevent spine enlargement, such as calcium channel blockers, N‐methyl‐D‐aspartate receptor antagonists, alpha‐amino‐2,3‐dihydro‐5‐methyl‐3‐oxo‐4‐isoxazolepropanoic acid receptor antagonists, and metabotropic glutamate receptor antagonists, are candidates for treatment of levodopa‐induced complications in PD. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Haruo Nishijima
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tatsuya Ueno
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yukihisa Funamizu
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Shinya Ueno
- Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
36
|
Villalba RM, Smith Y. Loss and remodeling of striatal dendritic spines in Parkinson's disease: from homeostasis to maladaptive plasticity? J Neural Transm (Vienna) 2017; 125:431-447. [PMID: 28540422 DOI: 10.1007/s00702-017-1735-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
In Parkinson's disease (PD) patients and animal models of PD, the progressive degeneration of the nigrostriatal dopamine (DA) projection leads to two major changes in the morphology of striatal projection neurons (SPNs), i.e., a profound loss of dendritic spines and the remodeling of axospinous glutamatergic synapses. Striatal spine loss is an early event tightly associated with the extent of striatal DA denervation, but not the severity of parkinsonian motor symptoms, suggesting that striatal spine pruning might be a form of homeostatic plasticity that compensates for the loss of striatal DA innervation and the resulting dysregulation of corticostriatal glutamatergic transmission. On the other hand, the remodeling of axospinous corticostriatal and thalamostriatal glutamatergic synapses might represent a form of late maladaptive plasticity that underlies changes in the strength and plastic properties of these afferents and the resulting increased firing and bursting activity of striatal SPNs in the parkinsonian state. There is also evidence that these abnormal synaptic connections might contribute to the pathophysiology of L-DOPA-induced dyskinesia. Despite the significant advances made in this field over the last thirty years, many controversial issues remain about the striatal SPN subtypes affected, the role of spine changes in the altered activity of SPNs in the parkinsonisn state, and the importance of striatal spine plasticity in the pathophysiology of L-DOPA-induced dyskinesia. In this review, we will examine the current state of knowledge of these issues, discuss the limitations of the animal models used to address some of these questions, and assess the relevance of data from animal models to the human-diseased condition.
Collapse
Affiliation(s)
- Rosa M Villalba
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA. .,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.,Department of Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
37
|
Andersen AD, Blaabjerg M, Binzer M, Kamal A, Thagesen H, Kjaer TW, Stenager E, Gramsbergen JBP. Cerebrospinal fluid levels of catecholamines and its metabolites in Parkinson's disease: effect of l-DOPA treatment and changes in levodopa-induced dyskinesia. J Neurochem 2017; 141:614-625. [PMID: 28244186 DOI: 10.1111/jnc.13997] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 11/28/2022]
Abstract
Levodopa (l-DOPA, l-3,4-dihydroxyphenylalanine) is the most effective drug in the symptomatic treatment of Parkinson's disease (PD), but chronic use initiates a maladaptive process leading to l-DOPA-induced dyskinesia (LID). Risk factors for early onset LID include younger age, more severe disease at baseline and higher daily l-DOPA dose, but biomarkers to predict the risk of motor complications are not yet available. Here, we investigated whether CSF levels of catecholamines and its metabolites are altered in PD patients with LID [PD-LID, n = 8)] as compared to non-dyskinetic PD patients receiving l-DOPA (PD-L, n = 6), or not receiving l-DOPA (PD-N, n = 7) as well as non-PD controls (n = 16). PD patients were clinically assessed using the Unified Parkinson's Disease Rating Scale and Unified Dyskinesia Rating Scale and CSF was collected after overnight fasting and 1-2 h after oral intake of l-DOPA or other anti-Parkinson medication. CSF catecholamines and its metabolites were analyzed by HPLC with electrochemical detection. We observed (i) decreased levels of dihydroxyphenylacetic acid (DOPAC) and homovanillic acid in PD patients not receiving l-DOPA (ii) higher dopamine (DA) levels in PD-LID as compared to controls (iii) higher DA/l-DOPA and lower DOPAC/DA ratio's in PD-LID as compared to PD-L and (iv) an age-dependent increase of DA and decrease of DOPAC/DA ratio in controls. These results suggest increased DA release from non-DA cells and deficient DA re-uptake in PD-LID. Monitoring DA and DOPAC in CSF of l-DOPA-treated PD patients may help identify patients at risk of developing LID.
Collapse
Affiliation(s)
- Andreas Dammann Andersen
- Department of Neurology, Hospital of Southern Jutland, Sønderborg, Denmark.,Institute of Regional Health Research, Center of Southern Jutland, University of Southern Denmark, Aabenraa, Denmark.,Focused Research Group in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark.,Odense Patient data Exploratory Network, Odense University Hospital, Odense, Denmark
| | - Morten Blaabjerg
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Department of Neurology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Michael Binzer
- Institute of Regional Health Research, Center of Southern Jutland, University of Southern Denmark, Aabenraa, Denmark.,Focused Research Group in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark
| | - Akram Kamal
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | - Helle Thagesen
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | | | - Egon Stenager
- Department of Neurology, Hospital of Southern Jutland, Sønderborg, Denmark.,Institute of Regional Health Research, Center of Southern Jutland, University of Southern Denmark, Aabenraa, Denmark.,Focused Research Group in Neurology, Hospital of Southern Jutland, Aabenraa, Denmark.,The Multiple Sclerosis Clinic of Southern Jutland, Vejle, Sonderborg, Esbjerg, Denmark
| | - Jan Bert Paul Gramsbergen
- Institute of Molecular Medicine, Neurobiological Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
38
|
Solís O, García-Montes JR, Garcia-Sanz P, Herranz AS, Asensio MJ, Kang G, Hiroi N, Moratalla R. Human COMT over-expression confers a heightened susceptibility to dyskinesia in mice. Neurobiol Dis 2017; 102:133-139. [PMID: 28315782 DOI: 10.1016/j.nbd.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 10/20/2022] Open
Abstract
Catechol-O-methyltransferase (COMT) degrades dopamine and its precursor l-DOPA and plays a critical role in regulating synaptic dopamine actions. We investigated the effects of heightened levels of COMT on dopamine-regulated motor behaviors and molecular alterations in a mouse model of dyskinesia. Transgenic mice overexpressing human COMT (TG) and their wildtype (WT) littermates received unilateral 6-OHDA lesions in the dorsal striatum and were treated chronically with l-DOPA for two weeks. l-DOPA-induced dyskinesia was exacerbated in TG mice without altering l-DOPA motor efficacy as determined by contralateral rotations or motor coordination. Inductions of FosB and phospho-acetylated histone 3 (molecular correlates of dyskinesia) were potentiated in the lesioned striatum of TG mice compared with their WT littermates. The TG mice had lower basal levels of dopamine in the striatum. In mice with lesions, l-DOPA induces a greater increase in the dopamine metabolite 3-methoxytyramine in the lesioned striatum of dyskinetic TG mice than in WT mice. The levels of serotonin and its metabolite were similar in TG and WT mice. Our results demonstrate that human COMT overexpression confers a heightened susceptibility to l-DOPA-induced dyskinesia and alters molecular and neurochemical responses in the lesioned striatum of mice.
Collapse
Affiliation(s)
- Oscar Solís
- Instituto Cajal, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose-Rubén García-Montes
- Instituto Cajal, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Garcia-Sanz
- Instituto Cajal, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio S Herranz
- Servicio de Neurobiología, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid 28034, Spain
| | - Maria-José Asensio
- Servicio de Neurobiología, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid 28034, Spain
| | - Gina Kang
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Noboru Hiroi
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rosario Moratalla
- Instituto Cajal, CSIC, Madrid 28002, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
39
|
Perez XA, Zhang D, Bordia T, Quik M. Striatal D1 medium spiny neuron activation induces dyskinesias in parkinsonian mice. Mov Disord 2017; 32:538-548. [PMID: 28256010 DOI: 10.1002/mds.26955] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/21/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Dyskinesias are a disabling motor complication that arises with prolonged l-dopa treatment. Studies using D1 receptor drugs and genetically modified mice suggest that medium spiny neurons expressing D1 receptors play a primary role in l-dopa-induced dyskinesias. However, the specific role of these neurons in dyskinesias is not fully understood. METHODS We used optogenetics, which allows for precise modulation of select neurons in vivo, to investigate whether striatal D1-expressing medium spiny neuron activity regulates abnormal involuntary movements or dyskinesia in parkinsonian mice. D1-cre mice unilaterally lesioned with 6-hydroxydopamine received striatal injections of cre-dependent channelrhodopsin2 virus or control virus. After stable virus expression, the effect of optical stimulation on dyskinesia was tested in l-dopa-naïve and l-dopa-primed mice. RESULTS Single-pulse and burst-optical stimulation of D1-expressing medium spiny neurons induced dyskinesias in l-dopa-naïve channelrhodopsin2 mice. In stably dyskinetic mice, l-dopa injection induced dyskinesia to a similar or somewhat greater extent than optical stimulation. Combined l-dopa administration and stimulation resulted in an additive increase in dyskinesias, indicating that other mechanisms also contribute. Molecular studies indicate that changes in extracellular signal-regulated kinase phosphorylation in D1-expressing medium spiny neurons are involved. Optical stimulation did not ameliorate parkinsonism in l-dopa-naïve mice. However, it improved parkinsonism in l-dopa-primed mice to a similar extent as l-dopa administration. None of the stimulation paradigms enhanced dyskinesia or modified parkinsonism in l-dopa-naïve or l-dopa-primed control virus mice. CONCLUSION The data provide direct evidence that striatal D1-expressing medium spiny neuron stimulation is sufficient to induce dyskinesias and contributes to the regulation of motor control. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xiomara A Perez
- Bioscience Division, SRI International, Menlo Park, California, USA
| | - Danhui Zhang
- Bioscience Division, SRI International, Menlo Park, California, USA
| | - Tanuja Bordia
- Bioscience Division, SRI International, Menlo Park, California, USA
| | - Maryka Quik
- Bioscience Division, SRI International, Menlo Park, California, USA
| |
Collapse
|
40
|
Alcacer C, Andreoli L, Sebastianutto I, Jakobsson J, Fieblinger T, Cenci MA. Chemogenetic stimulation of striatal projection neurons modulates responses to Parkinson's disease therapy. J Clin Invest 2017; 127:720-734. [PMID: 28112685 DOI: 10.1172/jci90132] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/22/2016] [Indexed: 01/14/2023] Open
Abstract
Parkinson's disease (PD) patients experience loss of normal motor function (hypokinesia), but can develop uncontrollable movements known as dyskinesia upon treatment with L-DOPA. Poverty or excess of movement in PD has been attributed to overactivity of striatal projection neurons forming either the indirect (iSPNs) or the direct (dSPNs) pathway, respectively. Here, we investigated the two pathways' contribution to different motor features using SPN type-specific chemogenetic stimulation in rodent models of PD (PD mice) and L-DOPA-induced dyskinesia (LID mice). Using the activatory Gq-coupled human M3 muscarinic receptor (hM3Dq), we found that chemogenetic stimulation of dSPNs mimicked, while stimulation of iSPNs abolished the therapeutic action of L-DOPA in PD mice. In LID mice, hM3Dq stimulation of dSPNs exacerbated dyskinetic responses to L-DOPA, while stimulation of iSPNs inhibited these responses. In the absence of L-DOPA, only chemogenetic stimulation of dSPNs mediated through the Gs-coupled modified rat muscarinic M3 receptor (rM3Ds) induced appreciable dyskinesia in PD mice. Combining D2 receptor agonist treatment with rM3Ds-dSPN stimulation reproduced all symptoms of LID. These results demonstrate that dSPNs and iSPNs oppositely modulate both therapeutic and dyskinetic responses to dopamine replacement therapy in PD. We also show that chemogenetic stimulation of different signaling pathways in dSPNs leads to markedly different motor outcomes. Our findings have important implications for the design of effective antiparkinsonian and antidyskinetic drug therapies.
Collapse
MESH Headings
- Animals
- Humans
- Levodopa/adverse effects
- Levodopa/pharmacology
- Mice
- Mice, Transgenic
- Neural Pathways/metabolism
- Neural Pathways/pathology
- Neurons/metabolism
- Neurons/pathology
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/drug therapy
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/pathology
- Rats
- Receptor, Muscarinic M3/agonists
- Receptor, Muscarinic M3/genetics
- Receptor, Muscarinic M3/metabolism
- Receptors, Dopamine D2/agonists
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Visual Cortex/metabolism
- Visual Cortex/pathology
Collapse
|
41
|
Nishi A, Shuto T. Potential for targeting dopamine/DARPP-32 signaling in neuropsychiatric and neurodegenerative disorders. Expert Opin Ther Targets 2017; 21:259-272. [PMID: 28052701 DOI: 10.1080/14728222.2017.1279149] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Alterations in dopamine neurotransmission has been implicated in pathophysiology of neuropsychiatric and neurodegenerative disorders, and DARPP-32 plays a pivotal role in dopamine neurotransmission. DARPP-32 likely influences dopamine-mediated behaviors in animal models of neuropsychiatric and neurodegenerative disorders and therapeutic effects of pharmacological treatment. Areas covered: We will review animal studies on the biochemical and behavioral roles of DARPP-32 in drug addiction, schizophrenia and Parkinson's disease. In general, under physiological and pathophysiological conditions, DARPP-32 in D1 receptor expressing (D1R) -medium spiny neurons (MSNs) promotes dopamine/D1 receptor/PKA signaling, whereas DARPP-32 in D2 receptor expressing (D2R)-MSNs counteracts dopamine/D2 receptor signaling. However, the function of DARPP-32 is differentially regulated in acute and chronic phases of drug addiction; DARPP-32 enhances D1 receptor/PKA signaling in the acute phase, whereas DARPP-32 suppresses D1 receptor/PKA signaling in the chronic phase through homeostatic mechanisms. Therefore, DARPP-32 plays a bidirectional role in dopamine neurotransmission, depending on the cell type and experimental conditions, and is involved in dopamine-related behavioral abnormalities. Expert opinion: DARPP-32 differentially regulates dopamine signaling in D1R- and D2R-MSNs, and a shift of balance between D1R- and D2R-MSN function is associated with behavioral abnormalities. An adjustment of this imbalance is achieved by therapeutic approaches targeting DARPP-32-related signaling molecules.
Collapse
Affiliation(s)
- Akinori Nishi
- a Department of Pharmacology , Kurume University School of Medicine , Kurume, Fukuoka , Japan
| | - Takahide Shuto
- a Department of Pharmacology , Kurume University School of Medicine , Kurume, Fukuoka , Japan
| |
Collapse
|
42
|
Bez F, Francardo V, Cenci MA. Dramatic differences in susceptibility to l-DOPA-induced dyskinesia between mice that are aged before or after a nigrostriatal dopamine lesion. Neurobiol Dis 2016; 94:213-25. [DOI: 10.1016/j.nbd.2016.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 12/26/2022] Open
|
43
|
Figge DA, Eskow Jaunarajs KL, Standaert DG. Dynamic DNA Methylation Regulates Levodopa-Induced Dyskinesia. J Neurosci 2016; 36:6514-24. [PMID: 27307239 PMCID: PMC5015786 DOI: 10.1523/jneurosci.0683-16.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/25/2016] [Accepted: 05/11/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Levodopa-induced dyskinesia (LID) is a persistent behavioral sensitization that develops after repeated levodopa (l-DOPA) exposure in Parkinson disease patients. LID is a consequence of sustained changes in the transcriptional behavior of striatal neurons following dopaminergic stimulation. In neurons, transcriptional regulation through dynamic DNA methylation has been shown pivotal to many long-term behavioral modifications; however, its role in LID has not yet been explored. Using a rodent model, we show LID development leads to the aberrant expression of DNA demethylating enzymes and locus-specific changes to DNA methylation at the promoter regions of genes aberrantly transcribed following l-DOPA treatment. Looking for dynamic DNA methylation in LID genome-wide, we used reduced representation bisulfite sequencing and found an extensive reorganization of the dorsal striatal methylome. LID development led to significant demethylation at many important regulatory areas of aberrantly transcribed genes. We used pharmacologic treatments that alter DNA methylation bidirectionally and found them able to modulate dyskinetic behaviors. Together, these findings demonstrate that l-DOPA induces widespread changes to striatal DNA methylation and that these modifications are required for the development and maintenance of LID. SIGNIFICANCE STATEMENT Levodopa-induced dyskinesia (LID) develops after repeated levodopa (l-DOPA) exposure in Parkinson disease patients and remains one of the primary obstacles to effective treatment. LID behaviors are a consequence of striatal neuron sensitization due to sustained changes in transcriptional behavior; however, the mechanisms responsible for the long-term maintenance of this cellular priming remain uncertain. Regulation of dynamic DNA methylation has been shown pivotal to the maintenance of several long-term behavioral modifications, yet its role in LID has not yet been explored. In this work, we report a pivotal role for the reorganization of DNA methylation in the development of LID and show that modification of DNA methylation may be a novel therapeutic target for use in preventing or reversing dyskinetic behaviors.
Collapse
Affiliation(s)
- David A Figge
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Karen L Eskow Jaunarajs
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - David G Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
44
|
Gurevich EV, Gainetdinov RR, Gurevich VV. G protein-coupled receptor kinases as regulators of dopamine receptor functions. Pharmacol Res 2016; 111:1-16. [PMID: 27178731 DOI: 10.1016/j.phrs.2016.05.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/03/2016] [Accepted: 05/06/2016] [Indexed: 02/08/2023]
Abstract
Actions of the neurotransmitter dopamine in the brain are mediated by dopamine receptors that belong to the superfamily of G protein-coupled receptors (GPCRs). Mammals have five dopamine receptor subtypes, D1 through D5. D1 and D5 couple to Gs/olf and activate adenylyl cyclase, whereas D2, D3, and D4 couple to Gi/o and inhibit it. Most GPCRs upon activation by an agonist are phosphorylated by GPCR kinases (GRKs). The GRK phosphorylation makes receptors high-affinity binding partners for arrestin proteins. Arrestin binding to active phosphorylated receptors stops further G protein activation and promotes receptor internalization, recycling or degradation, thereby regulating their signaling and trafficking. Four non- visual GRKs are expressed in striatal neurons. Here we describe known effects of individual GRKs on dopamine receptors in cell culture and in the two in vivo models of dopamine-mediated signaling: behavioral response to psychostimulants and L-DOPA- induced dyskinesia. Dyskinesia, associated with dopamine super-sensitivity of striatal neurons, is a debilitating side effect of L-DOPA therapy in Parkinson's disease. In vivo, GRK subtypes show greater receptor specificity than in vitro or in cultured cells. Overexpression, knockdown, and knockout of individual GRKs, particularly GRK2 and GRK6, have differential effects on signaling of dopamine receptor subtypes in the brain. Furthermore, deletion of GRK isoforms in select striatal neuronal types differentially affects psychostimulant-induced behaviors. In addition, anti-dyskinetic effect of GRK3 does not require its kinase activity: it is mediated by the binding of its RGS-like domain to Gαq/11, which suppresses Gq/11 signaling. The data demonstrate that the dopamine signaling in defined neuronal types in vivo is regulated by specific and finely orchestrated actions of GRK isoforms.
Collapse
Affiliation(s)
- Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37221, USA.
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; Skolkovo Institute of Science and Technology, Skolkovo, 143025, Moscow, Russia
| | | |
Collapse
|
45
|
Aristieta A, Ruiz-Ortega J, Miguelez C, Morera-Herreras T, Ugedo L. Chronic L-DOPA administration increases the firing rate but does not reverse enhanced slow frequency oscillatory activity and synchronization in substantia nigra pars reticulata neurons from 6-hydroxydopamine-lesioned rats. Neurobiol Dis 2016; 89:88-100. [DOI: 10.1016/j.nbd.2016.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 12/18/2022] Open
|
46
|
Effect of the C-terminal domain of the heavy chain of tetanus toxin on dyskinesia caused by levodopa in 6-hydroxydopamine-lesioned rats. Pharmacol Biochem Behav 2016; 145:33-44. [PMID: 27090294 DOI: 10.1016/j.pbb.2016.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 11/24/2022]
|
47
|
Solís O, García-Sanz P, Herranz AS, Asensio MJ, Moratalla R. L-DOPA Reverses the Increased Free Amino Acids Tissue Levels Induced by Dopamine Depletion and Rises GABA and Tyrosine in the Striatum. Neurotox Res 2016; 30:67-75. [PMID: 26966009 DOI: 10.1007/s12640-016-9612-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/30/2022]
Abstract
Perturbations in the cerebral levels of various amino acids are associated with neurological disorders, and previous studies have suggested that such alterations have a role in the motor and non-motor symptoms of Parkinson's disease. However, the direct effects of chronic L-DOPA treatment, that produces dyskinesia, on neural tissue amino acid concentrations have not been explored in detail. To evaluate whether striatal amino acid concentrations are altered in peak dose dyskinesia, 6-hydroxydopamine (6-OHDA)-lesioned hemiparkinsonian mice were treated chronically with L-DOPA and tissue amino acid concentrations were assessed by HPLC analysis. These experiments revealed that neither 6-OHDA nor L-DOPA treatment are able to alter glutamate in the striatum. However, glutamine increases after 6-OHDA and returns back to normal levels with L-DOPA treatment, suggesting increased striatal glutamatergic transmission with lack of dopamine. In addition, glycine and taurine levels are increased following dopamine denervation and restored to normal levels by L-DOPA. Interestingly, dyskinetic animals showed increased levels of GABA and tyrosine, while aspartate striatal tissue levels are not altered. Overall, our results indicate that chronic L-DOPA treatment, besides normalizing the altered levels of some amino acids after 6-OHDA, robustly increases striatal GABA and tyrosine levels which may in turn contribute to the development of L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Oscar Solís
- Instituto Cajal, CSIC, Av. Dr. Arce 37, 28002, Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia García-Sanz
- Instituto Cajal, CSIC, Av. Dr. Arce 37, 28002, Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio S Herranz
- Servicio Neurobiología, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - María-José Asensio
- Servicio Neurobiología, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Rosario Moratalla
- Instituto Cajal, CSIC, Av. Dr. Arce 37, 28002, Madrid, Spain. .,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Gangarossa G, Guzman M, Prado VF, Prado MA, Daumas S, El Mestikawy S, Valjent E. Role of the atypical vesicular glutamate transporter VGLUT3 in l-DOPA-induced dyskinesia. Neurobiol Dis 2016; 87:69-79. [DOI: 10.1016/j.nbd.2015.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/14/2015] [Accepted: 12/18/2015] [Indexed: 10/22/2022] Open
|
49
|
Bortolanza M, Padovan-Neto FE, Cavalcanti-Kiwiatkoski R, Dos Santos-Pereira M, Mitkovski M, Raisman-Vozari R, Del-Bel E. Are cyclooxygenase-2 and nitric oxide involved in the dyskinesia of Parkinson's disease induced by L-DOPA? Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0190. [PMID: 26009769 DOI: 10.1098/rstb.2014.0190] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Inflammatory mechanisms are proposed to play a role in L-DOPA-induced dyskinesia. Cyclooxygenase-2 (COX2) contributes to inflammation pathways in the periphery and is constitutively expressed in the central nervous system. Considering that inhibition of nitric oxide (NO) formation attenuates L-DOPA-induced dyskinesia, this study aimed at investigating if a NO synthase (NOS) inhibitor would change COX2 brain expression in animals with L-DOPA-induced dyskinesia. To this aim, male Wistar rats received unilateral 6-hydroxydopamine microinjection into the medial forebrain bundle were treated daily with L-DOPA (21 days) combined with 7-nitroindazole or vehicle. All hemi-Parkinsonian rats receiving l-DOPA showed dyskinesia. They also presented increased neuronal COX2 immunoreactivity in the dopamine-depleted dorsal striatum that was directly correlated with dyskinesia severity. Striatal COX2 co-localized with choline-acetyltransferase, calbindin and DARPP-32 (dopamine-cAMP-regulated phosphoprotein-32), neuronal markers of GABAergic neurons. NOS inhibition prevented L-DOPA-induced dyskinesia and COX2 increased expression in the dorsal striatum. These results suggest that increased COX2 expression after L-DOPA long-term treatment in Parkinsonian-like rats could contribute to the development of dyskinesia.
Collapse
Affiliation(s)
- Mariza Bortolanza
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil
| | - Fernando E Padovan-Neto
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil
| | - Roberta Cavalcanti-Kiwiatkoski
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Maurício Dos Santos-Pereira
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Miso Mitkovski
- Light Microscopy Facility, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Rita Raisman-Vozari
- Institut de Cerveau et de la Moelle Epinière, Sorbonne Université UPMC UM75 INSERM U1127, CNRS UMR 7225, Paris, France
| | - Elaine Del-Bel
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
50
|
Gurevich EV, Gainetdinov RR, Gurevich VV. Regulation of Dopamine-Dependent Behaviors by G Protein-Coupled Receptor Kinases. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3798-1_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|