1
|
Tufvesson-Alm M, Aranäs C, Blid Sköldheden S, Vestlund J, Edvardsson CE, Jerlhag E. LEAP2, a ghrelin receptor inverse agonist, and its effect on alcohol-related responses in rodents. Transl Psychiatry 2024; 14:401. [PMID: 39358354 PMCID: PMC11446955 DOI: 10.1038/s41398-024-03136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The underlying neurobiology of alcohol use disorder (AUD) is complex and needs further unraveling, with one of the key mechanisms being the gut-brain peptide ghrelin and its receptor (GHSR). However, additional substrates of the ghrelin pathway, such as liver-expressed antimicrobial peptide 2 (LEAP2), an endogenous GHSR inverse agonist, may contribute to this neurobiological framework. While LEAP2 modulates feeding and reward through central mechanisms, its effects on alcohol responses are unknown. The aim of the present study was therefore to identify the impact of central LEAP2 on the ability of alcohol to activate the mesolimbic dopamine system and to define its ability to control alcohol intake. These experiments revealed that central LEAP2 (i.e. into the third ventricle) prevented the ability of alcohol to cause locomotor stimulation in male mice, suppressed the memory of alcohol reward and attenuated the dopamine release in the nucleus accumbens caused by alcohol. Moreover, central LEAP2 reduced alcohol consumption in both male and female rats exposed to alcohol for 6 weeks before treatment. However, the serum levels of LEAP2 were similar between high- and low- alcohol-consuming (male) rats. Furthermore, central LEAP2 lowered the food intake in the alcohol-consuming male rats and reduced the body weight in the females. Collectively, the present study revealed that central LEAP2 mitigates alcohol-related responses in rodents, contributing to our understanding of the ghrelin pathway's role in alcohol effects.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
2
|
Sosero YL, Bandres-Ciga S, Ferwerda B, Tocino MTP, Belloso DR, Gómez-Garre P, Faouzi J, Taba P, Pavelka L, Marques TM, Gomes CPC, Kolodkin A, May P, Milanowski LM, Wszolek ZK, Uitti RJ, Heutink P, van Hilten JJ, Simon DK, Eberly S, Alvarez I, Krohn L, Yu E, Freeman K, Rudakou U, Ruskey JA, Asayesh F, Menéndez-Gonzàlez M, Pastor P, Ross OA, Krüger R, Corvol JC, Koks S, Mir P, De Bie RM, Iwaki H, Gan-Or Z. Dopamine Pathway and Parkinson's Risk Variants Are Associated with Levodopa-Induced Dyskinesia. Mov Disord 2024; 39:1773-1783. [PMID: 39132902 PMCID: PMC11490412 DOI: 10.1002/mds.29960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Levodopa-induced dyskinesia (LID) is a common adverse effect of levodopa, one of the main therapeutics used to treat the motor symptoms of Parkinson's disease (PD). Previous evidence suggests a connection between LID and a disruption of the dopaminergic system as well as genes implicated in PD, including GBA1 and LRRK2. OBJECTIVES Our goal was to investigate the effects of genetic variants on risk and time to LID. METHODS We performed a genome-wide association study (GWAS) and analyses focused on GBA1 and LRRK2 variants. We also calculated polygenic risk scores (PRS) including risk variants for PD and variants in genes involved in the dopaminergic transmission pathway. To test the influence of genetics on LID risk we used logistic regression, and to examine its impact on time to LID we performed Cox regression including 1612 PD patients with and 3175 without LID. RESULTS We found that GBA1 variants were associated with LID risk (odds ratio [OR] = 1.65; 95% confidence interval [CI], 1.21-2.26; P = 0.0017) and LRRK2 variants with reduced time to LID onset (hazard ratio [HR] = 1.42; 95% CI, 1.09-1.84; P = 0.0098). The fourth quartile of the PD PRS was associated with increased LID risk (ORfourth_quartile = 1.27; 95% CI, 1.03-1.56; P = 0.0210). The third and fourth dopamine pathway PRS quartiles were associated with a reduced time to development of LID (HRthird_quartile = 1.38; 95% CI, 1.07-1.79; P = 0.0128; HRfourth_quartile = 1.38; 95% CI = 1.06-1.78; P = 0.0147). CONCLUSIONS This study suggests that variants implicated in PD and in the dopaminergic transmission pathway play a role in the risk/time to develop LID. Further studies will be necessary to examine how these findings can inform clinical care. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yuri L. Sosero
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes on Health, Bethesda, MD, USA
| | - Bart Ferwerda
- Department of Clinical Epidemiology and Biostatistics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Maria T. P. Tocino
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Dìaz R. Belloso
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Johann Faouzi
- Sorbonne Université, Paris Brain Institute – ICM, Inserm, CNRS, Assistance Publique Hôpitaux de Paris, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal – BP 37203 35172 Bruz Cedex, France
| | - Pille Taba
- Department of Neurology and Neurosurgery, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Lukas Pavelka
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Tainà M. Marques
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Clarissa P. C. Gomes
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexey Kolodkin
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Patrick May
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
| | - Lukasz M Milanowski
- Department of Neurology Faculty of Health Science, Medical University of Warsaw, Warsaw, Poland
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | | | - Ryan J. Uitti
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | | | | | - David K. Simon
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School
| | - Shirley Eberly
- Department of Biostatistics and Computational Biology at the University of Rochester School of Medicine and Dentistry
| | - Ignacio Alvarez
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Kathryn Freeman
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Jennifer A. Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Farnaz Asayesh
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Manuel Menéndez-Gonzàlez
- Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Calle Julián Clavería s/n, 33006 Oviedo, Spain
- Department of Neurology, Hospital Universitario Central de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida Roma s/n, 33011 Oviedo, Spain
| | - Pau Pastor
- Department of Neurology, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Owen A. Ross
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-Christophe Corvol
- CREST, ENSAI, Campus de Ker-Lann, 51 Rue Blaise Pascal – BP 37203 35172 Bruz Cedex, France
| | - Sulev Koks
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Rob M.A. De Bie
- Department of Neurology and Clinical Neurophysiology, Amsterdam University Medical Centers, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes on Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, District of Columbia, USA
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
3
|
Piedade de Souza T, Santana de Araújo G, Magalhães L, Cavalcante GC, Ribeiro-Dos-Santos A, Sena-Dos-Santos C, Silva CS, Eufraseo GL, de Freitas Escudeiro A, Soares-Souza GB, Santos-Lobato BL, Ribeiro-Dos-Santos Â. Unveiling differential gene co-expression networks and its effects on levodopa-induced dyskinesia. iScience 2024; 27:110835. [PMID: 39297167 PMCID: PMC11409023 DOI: 10.1016/j.isci.2024.110835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/25/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Levodopa-induced dyskinesia (LID) refers to involuntary motor movements of chronic use of levodopa in Parkinson's disease (PD) that negatively impact the overall well-being of people with this disease. The molecular mechanisms involved in LID were investigated through whole-blood transcriptomic analysis for differential gene expression and identification of new co-expression and differential co-expression networks. We found six differentially expressed genes in patients with LID, and 13 in patients without LID. We also identified 12 co-expressed genes exclusive to LID, and six exclusive hub genes involved in 23 gene-gene interactions in patients with LID. Convergently, we identified novel genes associated with PD and LID that play roles in mitochondrial dysfunction, dysregulation of lipid metabolism, and neuroinflammation. We observed significant changes in disease progression, consistent with previous findings of maladaptive plastic changes in the basal ganglia leading to the development of LID, including a chronic pro-inflammatory state in the brain.
Collapse
Affiliation(s)
- Tatiane Piedade de Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | | | | | - Giovanna C Cavalcante
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Arthur Ribeiro-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Camille Sena-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Caio Santos Silva
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Gracivane Lopes Eufraseo
- Laboratório de Neurologia Experimental, Universidade Federal do Pará, Belém 66073-000, Pará, Brazil
| | | | - Giordano Bruno Soares-Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
- Instituto Tecnológico Vale, Belém 66055-090, Pará, Brazil
| | | | - Ândrea Ribeiro-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará (UFPA), Belém 66073-005, Pará, Brazil
| |
Collapse
|
4
|
Cardinale A, de Iure A, Picconi B. Neuroinflammation and Dyskinesia: A Possible Causative Relationship? Brain Sci 2024; 14:514. [PMID: 38790492 PMCID: PMC11118841 DOI: 10.3390/brainsci14050514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
Levodopa (L-DOPA) treatment represents the gold standard therapy for Parkinson's disease (PD) patients. L-DOPA therapy shows many side effects, among them, L-DOPA-induced dyskinesias (LIDs) remain the most problematic. Several are the mechanisms underlying these processes: abnormal corticostriatal neurotransmission, pre- and post-synaptic neuronal events, changes in gene expression, and altered plasticity. In recent years, researchers have also suggested non-neuronal mechanisms as a possible cause for LIDs. We reviewed recent clinical and pre-clinical studies on neuroinflammation contribution to LIDs. Microglia and astrocytes seem to play a strategic role in LIDs phenomenon. In particular, their inflammatory response affects neuron-glia communication, synaptic activity and neuroplasticity, contributing to LIDs development. Finally, we describe possible new therapeutic interventions for dyskinesia prevention targeting glia cells.
Collapse
Affiliation(s)
- Antonella Cardinale
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Antonio de Iure
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Barbara Picconi
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.C.); (A.d.I.)
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, 00166 Rome, Italy
| |
Collapse
|
5
|
Tufvesson-Alm M, Zhang Q, Aranäs C, Blid Sköldheden S, Edvardsson CE, Jerlhag E. Decoding the influence of central LEAP2 on food intake and its effect on accumbal dopamine release. Prog Neurobiol 2024; 236:102615. [PMID: 38641041 DOI: 10.1016/j.pneurobio.2024.102615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
The gut-brain peptide ghrelin and its receptor are established as a regulator of hunger and reward-processing. However, the recently recognized ghrelin receptor inverse agonist, liver-expressed antimicrobial peptide 2 (LEAP2), is less characterized. The present study aimed to elucidate LEAP2s central effect on reward-related behaviors through feeding and its mechanism. LEAP2 was administrated centrally in mice and effectively reduced feeding and intake of palatable foods. Strikingly, LEAP2s effect on feeding was correlated to the preference of the palatable food. Further, LEAP2 reduced the rewarding memory of high preference foods, and attenuated the accumbal dopamine release associated with palatable food exposure and eating. Interestingly, LEAP2 was widely expressed in the brain, and particularly in reward-related brain areas such as the laterodorsal tegmental area (LDTg). This expression was markedly altered when allowed free access to palatable foods. Accordingly, infusion of LEAP2 into LDTg was sufficient to transiently reduce acute palatable food intake. Taken together, the present results show that central LEAP2 has a profound effect on dopaminergic reward signaling associated with food and affects several aspects of feeding. The present study highlights LEAP2s effect on reward, which may have applications for obesity and other reward-related psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Qian Zhang
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Sebastian Blid Sköldheden
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Christian E Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden.
| |
Collapse
|
6
|
Chen YH, Kuo TT, Wang V, Cheng PW, Huang EYK, Ma KH, Greig NH, Olson L, Hoffer BJ, Tseng KY. Serotonergic Regulation of Synaptic Dopamine Levels Mitigates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:941-964. [PMID: 38905058 PMCID: PMC11307072 DOI: 10.3233/jpd-240080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/23/2024]
Abstract
Background The serotonin (5-HT) system can manipulate the processing of exogenous L-DOPA in the DA-denervated striatum, resulting in the modulation of L-DOPA-induced dyskinesia (LID). Objective To characterize the effects of the serotonin precursor 5-hydroxy-tryptophan (5-HTP) or the serotonin transporter (SERT) inhibitor, Citalopram on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson's disease. Methods MitoPark (MP) mice at 20 weeks of age, subjected to a 14-day administration of L-DOPA/Carbidopa, displayed dyskinesia, referred to as LID. Subsequent investigations explored the effects of 5-HT-modifying agents, such as 5-HTP and Citalopram, on abnormal involuntary movements (AIMs), locomotor activity, neurochemical signals, serotonin transporter activity, and protein expression in the DA-denervated striatum of LID MP mice. Results 5-HTP exhibited duration-dependent suppressive effects on developing and established LID, especially related to abnormal limb movements observed in L-DOPA-primed MP mice. However, Citalopram, predominantly suppressed abnormal axial movement induced by L-DOPA in LID MP mice. We demonstrated that 5-HTP could decrease L-DOPA-upregulation of DA turnover rates while concurrently upregulating 5-HT metabolism. Additionally, 5-HTP was shown to reduce the expressions of p-ERK and p-DARPP-32 in the striatum of LID MP mice. The effect of Citalopram in alleviating LID development may be attributed to downregulation of SERT activity in the dorsal striatum of LID MP mice. Conclusions While both single injection of 5-HTP and Citalopram effectively mitigated the development of LID, the difference in mitigation of AIM subtypes may be linked to the unique effects of these two serotonergic agents on L-DOPA-derived DA and 5-HT metabolism.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Vicki Wang
- Doctoral Degree Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | - Pin-Wen Cheng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | | | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Barry J. Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Cleveland, OH, USA
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
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
|
8
|
Conti Mazza MM, Centner A, Werner DF, Bishop C. Striatal serotonin transporter gain-of-function in L-DOPA-treated, hemi-parkinsonian rats. Brain Res 2023; 1811:148381. [PMID: 37127174 PMCID: PMC10562932 DOI: 10.1016/j.brainres.2023.148381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
L-DOPA is the standard treatment for Parkinson's disease (PD), but chronic treatment typically leads to L-DOPA-induced dyskinesia (LID). LID involves a complex interaction between the remaining dopamine (DA) system and the semi-homologous serotonin (5-HT) system. Since serotonin transporters (SERT) have some affinity for DA uptake, they may serve as a functional compensatory mechanism when DA transporters (DAT) are scant. DAT and SERT's functional contributions in the dyskinetic brain have not been well delineated. The current investigation sought to determine how DA depletion and L-DOPA treatment affect DAT and SERT transcriptional processes, translational processes, and functional DA uptake in the 6-hydroxydopamine-lesioned hemi-parkinsonian rat. Rats were counterbalanced for motor impairment into equally lesioned treatment groups then given daily L-DOPA (0 or 6 mg/kg) for 2 weeks. At the end of treatment, the substantia nigra was processed for tyrosine hydroxylase (TH) and DAT gene expression and dorsal raphe was processed for SERT gene expression. The striatum was processed for synaptosomal DAT and SERT protein expression and ex vivo DA uptake. Nigrostriatal DA loss severely reduced DAT mRNA and protein expression in the striatum with minimal changes in SERT. L-DOPA treatment, while not significantly affecting DAT or SERT alone, did increase striatal SERT:DAT protein ratios. Using ex vivo microdialysis, L-DOPA treatment increased DA uptake via SERT when DAT was depleted. Overall, these results suggest that DA loss and L-DOPA treatment uniquely alter DAT and SERT, revealing implications for monoamine transporters as potential biomarkers and therapeutic targets in the hemi-parkinsonian model and dyskinetic PD patients.
Collapse
Affiliation(s)
- Melissa M Conti Mazza
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - Ashley Centner
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - David F Werner
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA.
| |
Collapse
|
9
|
Aranäs C, Edvardsson CE, Shevchouk OT, Zhang Q, Witley S, Blid Sköldheden S, Zentveld L, Vallöf D, Tufvesson-Alm M, Jerlhag E. Semaglutide reduces alcohol intake and relapse-like drinking in male and female rats. EBioMedicine 2023; 93:104642. [PMID: 37295046 PMCID: PMC10363436 DOI: 10.1016/j.ebiom.2023.104642] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Glucagon-like peptide1 receptor (GLP-1R) agonists have been found to reduce alcohol drinking in rodents and overweight patients with alcohol use disorder (AUD). However, the probability of low semaglutide doses, an agonist with higher potency and affinity for GLP-1R, to attenuate alcohol-related responses in rodents and the underlying neuronal mechanisms is unknown. METHODS In the intermittent access model, we examined the ability of semaglutide to decrease alcohol intake and block relapse-like drinking, as well as imaging the binding of fluorescently marked semaglutide to nucleus accumbens (NAc) in both male and female rats. The suppressive effect of semaglutide on alcohol-induced locomotor stimulation and in vivo dopamine release in NAc was tested in male mice. We evaluated effect of semaglutide on the in vivo release of dopamine metabolites (DOPAC and HVA) and gene expression of enzymes metabolising dopamine (MAOA and COMT) in male mice. FINDINGS In male and female rats, acute and repeated semaglutide administration reduced alcohol intake and prevented relapse-like drinking. Moreover, fluorescently labelled semaglutide was detected in NAc of alcohol-drinking male and female rats. Further, semaglutide attenuated the ability of alcohol to cause hyperlocomotion and to elevate dopamine in NAc in male mice. As further shown in male mice, semaglutide enhanced DOPAC and HVA in NAc when alcohol was onboard and increased the gene expression of COMT and MAOA. INTERPRETATION Altogether, this indicates that semaglutide reduces alcohol drinking behaviours, possibly via a reduction in alcohol-induced reward and NAc dependent mechanisms. As semaglutide also decreased body weight of alcohol-drinking rats of both sexes, upcoming clinical studies should test the plausibility that semaglutide reduces alcohol intake and body weight in overweight AUD patients. FUNDING Swedish Research Council (2019-01676), LUA/ALF (723941) from the Sahlgrenska University Hospital and the Swedish brain foundation.
Collapse
Affiliation(s)
- Cajsa Aranäs
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Qian Zhang
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sarah Witley
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lindsay Zentveld
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
10
|
Pinna A, Parekh P, Morelli M. Serotonin 5-HT 1A receptors and their interactions with adenosine A 2A receptors in Parkinson's disease and dyskinesia. Neuropharmacology 2023; 226:109411. [PMID: 36608814 DOI: 10.1016/j.neuropharm.2023.109411] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
The dopamine neuronal loss that characterizes Parkinson's Disease (PD) is associated to changes in neurotransmitters, such as serotonin and adenosine, which contribute to the symptomatology of PD and to the onset of dyskinetic movements associated to levodopa treatment. The present review describes the role played by serotonin 5-HT1A receptors and the adenosine A2A receptors on dyskinetic movements induced by chronic levodopa in PD. The focus is on preclinical and clinical results showing the interaction between serotonin 5-HT1A receptors and other receptors such as 5-HT1B receptors and adenosine A2A receptors. 5-HT1A/1B receptor agonists and A2A receptor antagonists, administered in combination, contrast dyskinetic movements induced by chronic levodopa without impairing motor behaviour, suggesting that this drug combination might be a useful therapeutic approach for counteracting the PD motor deficits and dyskinesia associated with chronic levodopa treatment. This article is part of the Special Issue on "The receptor-receptor interaction as a new target for therapy".
Collapse
Affiliation(s)
- Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, c/o Department of Biomedical Sciences, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy.
| | - Pathik Parekh
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy
| | - Micaela Morelli
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, c/o Department of Biomedical Sciences, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy; Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy.
| |
Collapse
|
11
|
Espa E, Song L, Skovgård K, Fanni S, Cenci MA. Dopamine Agonist Cotreatment Alters Neuroplasticity and Pharmacology of Levodopa-Induced Dyskinesia. Mov Disord 2023; 38:410-422. [PMID: 36656044 PMCID: PMC10114531 DOI: 10.1002/mds.29301] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Current models of levodopa (L-dopa)-induced dyskinesia (LID) are obtained by treating dopamine-depleted animals with L-dopa. However, patients with LID receive combination therapies that often include dopamine agonists. OBJECTIVE Using 6-hydroxydopamine-lesioned rats as a model, we aimed to establish whether an adjunct treatment with the D2/3 agonist ropinirole impacts on patterns of LID-related neuroplasticity and drug responses. METHODS Different regimens of L-dopa monotreatment and L-dopa-ropinirole cotreatment were compared using measures of hypokinesia and dyskinesia. Striatal expression of ∆FosB and angiogenesis markers were studied immunohistochemically. Antidyskinetic effects of different drug categories were investigated in parallel groups of rats receiving either L-dopa monotreatment or L-dopa combined with ropinirole. RESULTS We defined chronic regimens of L-dopa monotreatment and L-dopa-ropinirole cotreatment inducing overall similar abnormal involuntary movement scores. Compared with the monotreatment group, animals receiving the L-dopa-ropinirole combination exhibited an overall lower striatal expression of ∆FosB with a distinctive compartmental distribution. The expression of angiogenesis markers and blood-brain barrier hyperpermeability was markedly reduced after L-dopa-ropinirole cotreatment compared with L-dopa monotreatment. Moreover, significant group differences were detected upon examining the response to candidate antidyskinetic drugs. In particular, compounds modulating D1 receptor signaling had a stronger effect in the L-dopa-only group, whereas both amantadine and the selective NMDA antagonist MK801 produced a markedly larger antidyskinetic effect in L-dopa-ropinirole cotreated animals. CONCLUSIONS Cotreatment with ropinirole altered LID-related neuroplasticity and pharmacological response profiles. The impact of adjuvant dopamine agonist treatment should be taken into consideration when investigating LID mechanisms and candidate interventions in both clinical and experimental settings. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Elena Espa
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lu Song
- Department of Neurology, XinhuaHospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Katrine Skovgård
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Silvia Fanni
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M. Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
12
|
PT320, a Sustained-Release GLP-1 Receptor Agonist, Ameliorates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24054687. [PMID: 36902115 PMCID: PMC10002999 DOI: 10.3390/ijms24054687] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
To determine the efficacy of PT320 on L-DOPA-induced dyskinetic behaviors, and neurochemistry in a progressive Parkinson's disease (PD) MitoPark mouse model. To investigate the effects of PT320 on the manifestation of dyskinesia in L-DOPA-primed mice, a clinically translatable biweekly PT320 dose was administered starting at either 5 or 17-weeks-old mice. The early treatment group was given L-DOPA starting at 20 weeks of age and longitudinally evaluated up to 22 weeks. The late treatment group was given L-DOPA starting at 28 weeks of age and longitudinally observed up to 29 weeks. To explore dopaminergic transmission, fast scan cyclic voltammetry (FSCV) was utilized to measure presynaptic dopamine (DA) dynamics in striatal slices following drug treatments. Early administration of PT320 significantly mitigated the severity L-DOPA-induced abnormal involuntary movements; PT320 particularly improved excessive numbers of standing as well as abnormal paw movements, while it did not affect L-DOPA-induced locomotor hyperactivity. In contrast, late administration of PT320 did not attenuate any L-DOPA-induced dyskinesia measurements. Moreover, early treatment with PT320 was shown to not only increase tonic and phasic release of DA in striatal slices in L-DOPA-naïve MitoPark mice, but also in L-DOPA-primed animals. Early treatment with PT320 ameliorated L-DOPA-induced dyskinesia in MitoPark mice, which may be related to the progressive level of DA denervation in PD.
Collapse
|
13
|
Lipari N, Centner A, Glinski J, Cohen S, Manfredsson FP, Bishop C. Characterizing the relationship between L-DOPA-induced-dyskinesia and psychosis-like behaviors in a bilateral rat model of Parkinson's disease. Neurobiol Dis 2023; 176:105965. [PMID: 36526089 DOI: 10.1016/j.nbd.2022.105965] [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: 10/12/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease associated psychosis (PDAP) is a prevalent non-motor symptom (NMS) that significantly erodes patients' and caregivers' quality of life yet remains vastly understudied. One potential source of PDAP in late-stage Parkinson's disease (PD) is the common dopamine (DA) replacement therapy for motor symptoms, Levodopa (L-DOPA). Given the high incidence of L-DOPA-induced dyskinesia (LID) in later phases of PD, this study sought to characterize the relationship between PDAP and LID in a bilateral medial forebrain bundle 6-hydroxydopamine hydrobromide (6-OHDA) lesion rat model. To assess PDAP in this model, prepulse inhibition (PPI), a well-validated assay of sensorimotor gating, was employed. First, we tested whether a bilateral lesion alone or after chronic L-DOPA treatment was sufficient to induce PPI dysfunction. Rats were also monitored for LID development, using the abnormal involuntary movements (AIMs) test, to examine PPI and LID associations. In experiment 2, Vilazodone (VZD), a serotonin transporter (SERT) blocker and 1A receptor (5-HT1A) partial agonist was administered to test its potential efficacy in reducing LID and PPI dysfunction. Once testing was complete, tissue was collected for high performance liquid chromatography (HPLC) to examine the monoamine levels in motor and non-motor circuits. Results indicate that bilateral DA lesions produced motor deficits and that chronic L-DOPA induced moderate AIMs; importantly, rats that developed more severe AIMs were more likely to display sensorimotor gating dysfunction. In addition, VZD treatment dose-dependently reduced L-DOPA-induced AIMs without impairing L-DOPA efficacy, although VZD's effects on PPI were limited. Altogether, this project established the bilateral 6-OHDA lesion model accurately portrayed LID and PDAP-like behaviors, uncovered their potential relationship, and finally, demonstrated the utility of VZD for reducing LID.
Collapse
Affiliation(s)
- Natalie Lipari
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Ashley Centner
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - John Glinski
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Sophie Cohen
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | | | | |
Collapse
|
14
|
Skovgård K, Barrientos SA, Petersson P, Halje P, Cenci MA. Distinctive Effects of D1 and D2 Receptor Agonists on Cortico-Basal Ganglia Oscillations in a Rodent Model of L-DOPA-Induced Dyskinesia. Neurotherapeutics 2023; 20:304-324. [PMID: 36344723 PMCID: PMC10119363 DOI: 10.1007/s13311-022-01309-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 11/09/2022] Open
Abstract
L-DOPA-induced dyskinesia (LID) in Parkinson's disease has been linked to oscillatory neuronal activities in the cortico-basal ganglia network. We set out to examine the pattern of cortico-basal ganglia oscillations induced by selective agonists of D1 and D2 receptors in a rat model of LID. Local field potentials were recorded in freely moving rats using large-scale electrodes targeting three motor cortical regions, dorsomedial and dorsolateral striatum, external globus pallidus, and substantial nigra pars reticulata. Abnormal involuntary movements were elicited by the D1 agonist SKF82958 or the D2 agonist sumanirole, while overall motor activity was quantified using video analysis (DeepLabCut). Both SKF82958 and sumanirole induced dyskinesia, although with significant differences in temporal course, overall severity, and body distribution. The D1 agonist induced prominent narrowband oscillations in the high gamma range (70-110 Hz) in all recorded structures except for the nigra reticulata. Additionally, the D1 agonist induced strong functional connectivity between the recorded structures and the phase analysis revealed that the primary motor cortex (forelimb area) was leading a supplementary motor area and striatum. Following treatment with the D2 agonist, narrowband gamma oscillations were detected only in forelimb motor cortex and dorsolateral striatum, while prominent oscillations in the theta band occurred in the globus pallidus and nigra reticulata. Our results reveal that the dyskinetic effects of D1 and D2 receptor agonists are associated with distinct patterns of cortico-basal ganglia oscillations, suggesting a recruitment of partially distinct networks.
Collapse
Affiliation(s)
- Katrine Skovgård
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC A13, 221 84, Lund, Sweden
- The Group for Integrative Neurophysiology and Neurotechnology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sebastian A Barrientos
- The Group for Integrative Neurophysiology and Neurotechnology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Per Petersson
- The Group for Integrative Neurophysiology and Neurotechnology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Pär Halje
- The Group for Integrative Neurophysiology and Neurotechnology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC A13, 221 84, Lund, Sweden.
| |
Collapse
|
15
|
Levodopa-Induced Dyskinesia in Parkinson's Disease: Pathogenesis and Emerging Treatment Strategies. Cells 2022; 11:cells11233736. [PMID: 36496996 PMCID: PMC9736114 DOI: 10.3390/cells11233736] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
The most commonly used treatment for Parkinson's disease (PD) is levodopa, prescribed in conjunction with carbidopa. Virtually all patients with PD undergo dopamine replacement therapy using levodopa during the course of the disease's progression. However, despite the fact that levodopa is the "gold standard" in PD treatments and has the ability to significantly alleviate PD symptoms, it comes with side effects in advanced PD. Levodopa replacement therapy remains the current clinical treatment of choice for Parkinson's patients, but approximately 80% of the treated PD patients develop levodopa-induced dyskinesia (LID) in the advanced stages of the disease. A better understanding of the pathological mechanisms of LID and possible means of improvement would significantly improve the outcome of PD patients, reduce the complexity of medication use, and lower adverse effects, thus, improving the quality of life of patients and prolonging their life cycle. This review assesses the recent advancements in understanding the underlying mechanisms of LID and the therapeutic management options available after the emergence of LID in patients. We summarized the pathogenesis and the new treatments for LID-related PD and concluded that targeting pathways other than the dopaminergic pathway to treat LID has become a new possibility, and, currently, amantadine, drugs targeting 5-hydroxytryptamine receptors, and surgery for PD can target the Parkinson's symptoms caused by LID.
Collapse
|
16
|
Florio E, Serra M, Lewis RG, Kramár E, Freidberg M, Wood M, Morelli M, Borrelli E. D2R signaling in striatal spiny neurons modulates L-DOPA induced dyskinesia. iScience 2022; 25:105263. [PMID: 36274959 PMCID: PMC9579025 DOI: 10.1016/j.isci.2022.105263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/19/2022] [Accepted: 09/25/2022] [Indexed: 11/07/2022] Open
Abstract
Degeneration of dopaminergic neurons leads to Parkinson's disease (PD), characterized by reduced levels of striatal dopamine (DA) and impaired voluntary movements. DA replacement is achieved by levodopa treatment which in long-term causes involuntary movements or dyskinesia. Dyskinesia is linked to the pulsatile activation of D1 receptors of the striatal medium spiny neurons (MSNs) forming the direct output pathway (dMSNs). The contribution of DA stimulation of D2R in MSNs of the indirect pathway (iMSNs) is less clear. Using the 6-hydroxydopamine model of PD, here we show that loss of DA-mediated inhibition of these neurons intensifies levodopa-induced dyskinesia (LID) leading to reprogramming of striatal gene expression. We propose that the motor impairments characteristic of PD and of its therapy are critically dependent on D2R-mediated iMSNs activity. D2R signaling not only filters inputs to the striatum but also indirectly regulates dMSNs mediated responses.
Collapse
Affiliation(s)
- Ermanno Florio
- Department of Microbiology & Molecular Genetics, INSERM U1233, Center for Epigenetics and Metabolism, 308 Sprague Hall, University of California, Irvine, Irvine, CA 92697, USA
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato (CA), Italy
| | - Robert G. Lewis
- Department of Microbiology & Molecular Genetics, INSERM U1233, Center for Epigenetics and Metabolism, 308 Sprague Hall, University of California, Irvine, Irvine, CA 92697, USA
| | - Enikö Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, 200 Qureshey Research Lab., Irvine, CA 92697, USA
| | - Michael Freidberg
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences II, Irvine, CA 92697, USA
| | - Marcello Wood
- Department of Neurobiology and Behavior, University of California, Irvine, 200 Qureshey Research Lab., Irvine, CA 92697, USA
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato (CA), Italy
| | - Emiliana Borrelli
- Department of Microbiology & Molecular Genetics, INSERM U1233, Center for Epigenetics and Metabolism, 308 Sprague Hall, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Nishikawa N, Hatano T, Kamiyama D, Haginiwa-Hasegawa H, Oyama G, Hattori N. Continuous 24-h Levodopa-Carbidopa Intestinal Gel Infusion After a Levodopa Holiday Suppressed Refractory Dyskinesia Despite Increasing Levodopa Dose. J Mov Disord 2022; 15:290-292. [PMID: 35880379 PMCID: PMC9536906 DOI: 10.14802/jmd.22021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Noriko Nishikawa
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Daiki Kamiyama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Genko Oyama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Modulating Microglia/Macrophage Activation by CDNF Promotes Transplantation of Fetal Ventral Mesencephalic Graft Survival and Function in a Hemiparkinsonian Rat Model. Biomedicines 2022; 10:biomedicines10061446. [PMID: 35740467 PMCID: PMC9221078 DOI: 10.3390/biomedicines10061446] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in substantia nigra pars compacta, which leads to the motor control deficits. Recently, cell transplantation is a cutting-edge technique for the therapy of PD. Nevertheless, one key bottleneck to realizing such potential is allogenic immune reaction of tissue grafts by recipients. Cerebral dopamine neurotrophic factor (CDNF) was shown to possess immune-modulatory properties that benefit neurodegenerative diseases. We hypothesized that co-administration of CDNF with fetal ventral mesencephalic (VM) tissue can improve the success of VM replacement therapies by attenuating immune responses. Hemiparkinsonian rats were generated by injecting 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle of Sprague Dawley (SD) rats. The rats were then intrastriatally transplanted with VM tissue from rats, with/without CDNF administration. Recovery of dopaminergic function and survival of the grafts were evaluated using the apomorphine-induced rotation test and small-animal positron emission tomography (PET) coupled with [18F] DOPA or [18F] FE-PE2I, respectively. In addition, transplantation-related inflammatory response was determined by uptake of [18F] FEPPA in the grafted side of striatum. Immunohistochemistry (IHC) examination was used to determine the survival of the grated dopaminergic neurons in the striatum and to investigate immune-modulatory effects of CDNF. The modulation of inflammatory responses caused by CDNF might involve enhancing M2 subset polarization and increasing fractal dimensions of 6-OHDA-treated BV2 microglial cell line. Analysis of CDNF-induced changes to gene expressions of 6-OHDA-stimulated BV2 cells implies that these alternations of the biomarkers and microglial morphology are implicated in the upregulation of protein kinase B signaling as well as regulation of catalytic, transferase, and protein serine/threonine kinase activity. The effects of CDNF on 6-OHDA-induced alternation of the canonical pathway in BV2 microglial cells is highly associated with PI3K-mediated phagosome formation. Our results are the first to show that CDNF administration enhances the survival of the grafted dopaminergic neurons and improves functional recovery in PD animal model. Modulation of the polarization, morphological characteristics, and transcriptional profiles of 6-OHDA-stimualted microglia by CDNF may possess these properties in transplantation-based regenerative therapies.
Collapse
|
19
|
Cohen SR, Terry ML, Coyle M, Wheelis E, Centner A, Smith S, Glinski J, Lipari N, Budrow C, Manfredsson FP, Bishop C. The multimodal serotonin compound Vilazodone alone, but not combined with the glutamate antagonist Amantadine, reduces l-DOPA-induced dyskinesia in hemiparkinsonian rats. Pharmacol Biochem Behav 2022; 217:173393. [DOI: 10.1016/j.pbb.2022.173393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/06/2023]
|
20
|
Wilson L, Lee CA, Mason CF, Khodjaniyazova S, Flores KB, Muddiman DC, Sombers LA. Simultaneous Measurement of Striatal Dopamine and Hydrogen Peroxide Transients Associated with L-DOPA Induced Rotation in Hemiparkinsonian Rats. ACS MEASUREMENT SCIENCE AU 2022; 2:120-131. [PMID: 36785724 PMCID: PMC9838821 DOI: 10.1021/acsmeasuresciau.1c00030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder commonly treated with levodopa (L-DOPA), which eventually induces abnormal involuntary movements (AIMs). The neurochemical contributors to these dyskinesias are unknown; however, several lines of evidence indicate an interplay of dopamine (DA) and oxidative stress. Here, DA and hydrogen peroxide (H2O2) were simultaneously monitored at discrete recording sites in the dorsal striata of hemiparkinsonian rats using fast-scan cyclic voltammetry. Mass spectrometry imaging validated the lesions. Hemiparkinsonian rats exhibited classic L-DOPA-induced AIMs and rotations as well as increased DA and H2O2 tone over saline controls after 1 week of treatment. By week 3, DA tone remained elevated beyond that of controls, but H2O2 tone was largely normalized. At this time point, rapid chemical transients were time-locked with spontaneous bouts of rotation. Striatal H2O2 rapidly increased with the initiation of contraversive rotational behaviors in lesioned L-DOPA animals, in both hemispheres. DA signals simultaneously decreased with rotation onset. The results support a role for these striatal neuromodulators in the adaptive changes that occur with L-DOPA treatment in PD and reveal a precise interplay between DA and H2O2 in the initiation of involuntary locomotion.
Collapse
Affiliation(s)
- Leslie
R. Wilson
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - Christie A. Lee
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - Catherine F. Mason
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - Sitora Khodjaniyazova
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - Kevin B. Flores
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - David C. Muddiman
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| | - Leslie A. Sombers
- Department
of Chemistry, Department of Mathematics, Molecular Education, Technology,
and Research Innovation Center (METRIC), Center for Research in Scientific
Computation, and Comparative Medicine Institute, North Carolina
State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
21
|
Furukawa K, Shima A, Kambe D, Nishida A, Wada I, Sakamaki H, Yoshimura K, Terada Y, Sakato Y, Mitsuhashi M, Sawamura M, Nakanishi E, Taruno Y, Yamakado H, Fushimi Y, Okada T, Nakamoto Y, Takahashi R, Sawamoto N. Motor progression and nigrostriatal neurodegeneration in Parkinson’s disease. Ann Neurol 2022; 92:110-121. [DOI: 10.1002/ana.26373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/03/2022] [Accepted: 04/11/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Koji Furukawa
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Atsushi Shima
- Human Brain Research Center Kyoto University Graduate School of Medicine Kyoto Japan
| | - Daisuke Kambe
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Akira Nishida
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Ikko Wada
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Haruhi Sakamaki
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Kenji Yoshimura
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yuta Terada
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yusuke Sakato
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Masahiro Mitsuhashi
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Masanori Sawamura
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Etsuro Nakanishi
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yosuke Taruno
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Hodaka Yamakado
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Tomohisa Okada
- Human Brain Research Center Kyoto University Graduate School of Medicine Kyoto Japan
- Department of Diagnostic Imaging and Nuclear Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Ryosuke Takahashi
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Nobukatsu Sawamoto
- Department of Human Health Sciences Kyoto University Graduate School of Medicine Kyoto Japan
| |
Collapse
|
22
|
Bandopadhyay R, Mishra N, Rana R, Kaur G, Ghoneim MM, Alshehri S, Mustafa G, Ahmad J, Alhakamy NA, Mishra A. Molecular Mechanisms and Therapeutic Strategies for Levodopa-Induced Dyskinesia in Parkinson's Disease: A Perspective Through Preclinical and Clinical Evidence. Front Pharmacol 2022; 13:805388. [PMID: 35462934 PMCID: PMC9021725 DOI: 10.3389/fphar.2022.805388] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the second leading neurodegenerative disease that is characterized by severe locomotor abnormalities. Levodopa (L-DOPA) treatment has been considered a mainstay for the management of PD; however, its prolonged treatment is often associated with abnormal involuntary movements and results in L-DOPA-induced dyskinesia (LID). Although LID is encountered after chronic administration of L-DOPA, the appearance of dyskinesia after weeks or months of the L-DOPA treatment has complicated our understanding of its pathogenesis. Pathophysiology of LID is mainly associated with alteration of direct and indirect pathways of the cortico-basal ganglia-thalamic loop, which regulates normal fine motor movements. Hypersensitivity of dopamine receptors has been involved in the development of LID; moreover, these symptoms are worsened by concurrent non-dopaminergic innervations including glutamatergic, serotonergic, and peptidergic neurotransmission. The present study is focused on discussing the recent updates in molecular mechanisms and therapeutic approaches for the effective management of LID in PD patients.
Collapse
Affiliation(s)
- Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Nainshi Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ruhi Rana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Gagandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gulam Mustafa
- College of Pharmacy (Boys), Al-Dawadmi Campus, Shaqra University, Riyadh, Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Nabil. A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)—Guwahati, Guwahati, India
| |
Collapse
|
23
|
Hunter J, Bova A, Stevens A, Leventhal DK. Dopamine neuron stimulation induces context-dependent abnormal involuntary movements in healthy rats. iScience 2022; 25:103974. [PMID: 35281727 PMCID: PMC8914546 DOI: 10.1016/j.isci.2022.103974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/05/2022] [Accepted: 02/18/2022] [Indexed: 11/27/2022] Open
Abstract
With continued levodopa treatment, most patients with Parkinson disease (PD) develop levodopa-induced dyskinesias (LIDs)—abnormal involuntary movements (AIMs) characterized primarily by chorea. Clinically, LIDs depend on nigrostriatal degeneration and sensitization to repeated levodopa doses. However, the degree of dopamine denervation is correlated with levodopa-induced changes in striatal dopamine. Therefore, pulsatile dopamine release may induce AIMs independently of nigrostriatal degeneration. We optogenetically stimulated dopamine neurons in healthy rats as they engaged in skilled reaching. Repeated stimulation induced progressive AIMs whose severity was modified by behavioral context. AIMs were milder with stimulation during reaches, and more severe if stimulation occurred between reaches. Despite gradual induction, AIMs recurred immediately with subsequent dopamine neuron stimulation. Thus, nigrostriatal denervation is not necessary for fluctuating striatal dopamine to induce AIMs, and behavioral context modulates AIM expression. Furthermore, pulsatile dopamine release induces persistent changes in motor circuits that are revealed by subsequent dopamine neuron activation in appropriate contexts. Repeated dopamine neuron activation causes involuntary movements in healthy rats These movements resemble levodopa-induced dyskinesias in parkinsonian rats Movement severity depends on the history of prior stimulation Movement severity is diminished in rats actively engaged in goal-directed behavior
Collapse
Affiliation(s)
- Julia Hunter
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexandra Bova
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew Stevens
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel K Leventhal
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.,Parkinson Disease Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Neurology, VA Ann Arbor Health System, Ann Arbor, MI 48105, USA
| |
Collapse
|
24
|
Angela Cenci M, Skovgård K, Odin P. Non-dopaminergic approaches to the treatment of motor complications in Parkinson's disease. Neuropharmacology 2022; 210:109027. [DOI: 10.1016/j.neuropharm.2022.109027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/21/2022]
|
25
|
Gao S, Gao R, Yao L, Feng J, Liu W, Zhou Y, Zhang Q, Wang Y, Liu J. Striatal D1 Dopamine Neuronal Population Dynamics in a Rat Model of Levodopa-Induced Dyskinesia. Front Aging Neurosci 2022; 14:783893. [PMID: 35185524 PMCID: PMC8850470 DOI: 10.3389/fnagi.2022.783893] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/10/2022] [Indexed: 11/21/2022] Open
Abstract
Background The pathophysiology of levodopa-induced dyskinesia (LID) in Parkinson’s disease (PD) is not well understood. Experimental data from numerous investigations support the idea that aberrant activity of D1 dopamine receptor-positive medium spiny neurons in the striatal direct pathway is associated with LID. However, a direct link between the real-time activity of these striatal neurons and dyskinetic symptoms remains to be established. Methods We examined the effect of acute levodopa treatment on striatal c-Fos expression in LID using D1-Cre PD rats with dyskinetic symptoms induced by chronic levodopa administration. We studied the real-time dynamics of striatal D1+ neurons during dyskinetic behavior using GCaMP6-based in vivo fiber photometry. We also examined the effects of striatal D1+ neuronal deactivation on dyskinesia in LID rats using optogenetics and chemogenetic methods. Results Striatal D1+ neurons in LID rats showed increased expression of c-Fos, a widely used marker for neuronal activation, following levodopa injection. Fiber photometry revealed synchronized overactivity of striatal D1+ neurons during dyskinetic behavior in LID rats following levodopa administration. Consistent with these observations, optogenetic deactivation of striatal D1+ neurons was sufficient to inhibit most of the dyskinetic behaviors of LID animals. Moreover, chemogenetic inhibition of striatal D1+ neurons delayed the onset of dyskinetic behavior after levodopa administration. Conclusion Our data demonstrated that aberrant activity of striatal D1+ neuronal population was causally linked with real-time dyskinetic symptoms in LID rats.
Collapse
Affiliation(s)
- Shasha Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Rui Gao
- Department of Medical Imaging and Nuclear Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lu Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Jie Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wanyuan Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yingqiong Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Qiongchi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
- *Correspondence: Yong Wang,
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Xi’an Jiaotong University Health Science Center, Xi’an, China
| |
Collapse
|
26
|
Tseng KY, Kuo TT, Wang V, Huang EYK, Ma KH, Olson L, Hoffer BJ, Chen YH. Tetrabenazine Mitigates Aberrant Release and Clearance of Dopamine in the Nigrostriatal System, and Alleviates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1545-1565. [PMID: 35599497 DOI: 10.3233/jpd-223195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND L-DOPA-induced dyskinesia (LID), occurring with aberrant processing of exogenous L-DOPA in the dopamine-denervated striatum, is a main complication of levodopa treatment in Parkinson's disease. OBJECTIVE To characterize the effects of the vesicular antagonist tetrabenazine (TBZ) on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson's disease. METHODS 20-week-old MitoPark mice were co-treated or separately administered TBZ and L-DOPA for 14 days. Abnormal involuntary movements (AIMs) and locomotor activity were analyzed. To explore dopamine (DA) transmission, fast scan cyclic voltammetry was used to assess presynaptic DA dynamics in striatal slices following treatments. PET imaging with 4-[18F]-PE2I, ADAM and immunoblotting assays were used to detect receptor protein changes in the DA-denervated striatum. Finally, nigrostriatal tissues were collected for HPLC measures of DA, serotonin and their metabolites. RESULTS A single injection of TBZ given in the interval between the two L-DOPA/Carbidopa treatments significantly attenuated L-DOPA-induced AIMs expression and locomotor hyperactivity. TBZ was shown to reduce tonic and phasic release of DA following L-DOPA treatment in DA-denervated striatal tissue. In the DA-depleted striatum, TBZ decreased the expression of L-DOPA-enhanced D1 receptors and the serotonin reuptake transporter. Neurochemical analysis indicated that TBZ attenuated L-DOPA-induced surges of DA levels by promoting DA turnover in the nigrostriatal system. CONCLUSIONS Our findings demonstrate that TBZ diminishes abnormal striatal DA transmission, which involves the ability of TBZ to modulate the presymptomatic dynamics of DA, and then mitigate aberrant release of exogenous L-DOPA from nerve terminals. The results support the potential of repositioning TBZ to counteract LID development.
Collapse
Affiliation(s)
- Kuan-Yin Tseng
- National Defense Medical Center, Taipei, Taiwan, R.O.C
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| | - Vicki Wang
- Ph.D. Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yuan-Hao Chen
- National Defense Medical Center, Taipei, Taiwan, R.O.C
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
27
|
Soti M, Ranjbar H, Kohlmeier KA, Shabani M. Parkinson's disease related alterations in cannabinoid transmission. Brain Res Bull 2021; 178:82-96. [PMID: 34808322 DOI: 10.1016/j.brainresbull.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNc) by neurodegeneration. Recent findings in animal models of PD propose tonic inhibition of the remaining DA neurons through GABA release from reactive glial cells. Movement dysfunctions could be ameliorated by promotion of activity in dormant DA cells. The endocannabinoid system (ECS) is extensively present in basal ganglia (BG) and is known as an indirect modulator of DAergic neurotransmission, thus drugs designed to target this system have shown promising therapeutic potential in PD patients. Interestingly, down/up-regulation of cannabinoid receptors (CBRs) varies across the different stages of PD, suggesting that some of the motor/ non-motor deficits may be related to changes in CBRs. Determination of the profile of changes of these receptors across the different stages of PD as well as their neural distribution within the BG could improve understanding of PD and identify pathways important in disease pathobiology. In this review, we focus on temporal and spatial alterations of CBRs during PD in the BG. At present, as inconclusive, but suggestive results have been obtained, future investigations should be conducted to extend preclinical studies examining CBRs changes within each stage in controlled clinical trials in order to determine the potential of targeting CBRs in management of PD.
Collapse
Affiliation(s)
- Monavareh Soti
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Hoda Ranjbar
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
28
|
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
|
29
|
Vidal B, Levigoureux E, Chaib S, Bouillot C, Billard T, Newman-Tancredi A, Zimmer L. Different Alterations of Agonist and Antagonist Binding to 5-HT1A Receptor in a Rat Model of Parkinson’s Disease and Levodopa-Induced Dyskinesia: A MicroPET Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:1257-1269. [DOI: 10.3233/jpd-212580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: The gold-standard treatment for Parkinson’s disease is L-DOPA, which in the long term often leads to levodopa-induced dyskinesia. Serotonergic neurons are partially responsible for this, by converting L-DOPA into dopamine leading to its uncontrolled release as a “false neurotransmitter”. The stimulation of 5-HT1A receptors can reduce involuntary movements but this mechanism is poorly understood. Objective: This study aimed to investigate the functionality of 5-HT1A receptors using positron emission tomography in hemiparkinsonian rats with or without dyskinesia induced by 3-weeks daily treatment with L-DOPA. Imaging sessions were performed “off” L-DOPA. Methods: Each rat underwent a positron emission tomography scan with [18F]F13640, a 5-HT1AR agonist which labels receptors in a high affinity state for agonists, or with [18F]MPPF, a 5-HT1AR antagonist which labels all the receptors. Results: There were decreases of [18F]MPPF binding in hemiparkinsonian rats in cortical areas. In dyskinetic animals, changes were slighter but also found in other regions. In hemiparkinsonian rats, [18F]F13640 uptake was decreased bilaterally in the globus pallidus and thalamus. On the non-lesioned side, binding was increased in the insula, the hippocampus and the amygdala. In dyskinetic animals, [18F]F13640 binding was strongly increased in cortical and limbic areas, especially in the non-lesioned side. Conclusion: These data suggest that agonist and antagonist 5-HT1A receptor-binding sites are differently modified in Parkinson’s disease and levodopa-induced dyskinesia. In particular, these observations suggest a substantial involvement of the functional state of 5-HT1AR in levodopa-induced dyskinesia and emphasize the need to characterize this state using agonist radiotracers in physiological and pathological conditions.
Collapse
Affiliation(s)
- Benjamin Vidal
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
| | - Elise Levigoureux
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | - Sarah Chaib
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | | | - Thierry Billard
- CERMEP-Imaging Platform, Bron, France
- Institute of Chemistry and Biochemistry, Université de Lyon, CNRS, Villeurbanne, France
| | | | - Luc Zimmer
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
- CERMEP-Imaging Platform, Bron, France
| |
Collapse
|
30
|
Caligiore D, Montedori F, Buscaglione S, Capirchio A. Increasing Serotonin to Reduce Parkinsonian Tremor. Front Syst Neurosci 2021; 15:682990. [PMID: 34354572 PMCID: PMC8331097 DOI: 10.3389/fnsys.2021.682990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
While current dopamine-based drugs seem to be effective for most Parkinson's disease (PD) motor dysfunctions, they produce variable responsiveness for resting tremor. This lack of consistency could be explained by considering recent evidence suggesting that PD resting tremor can be divided into different partially overlapping phenotypes based on the dopamine response. These phenotypes may be associated with different pathophysiological mechanisms produced by a cortical-subcortical network involving even non-dopaminergic areas traditionally not directly related to PD. In this study, we propose a bio-constrained computational model to study the neural mechanisms underlying a possible type of PD tremor: the one mainly involving the serotoninergic system. The simulations run with the model demonstrate that a physiological serotonin increase can partially recover dopamine levels at the early stages of the disease before the manifestation of overt tremor. This result suggests that monitoring serotonin concentration changes could be critical for early diagnosis. The simulations also show the effectiveness of a new pharmacological treatment for tremor that acts on serotonin to recover dopamine levels. This latter result has been validated by reproducing existing data collected with human patients.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Francesco Montedori
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Silvia Buscaglione
- Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit (NeXT), Campus Bio-Medico University, Rome, Italy
| | - Adriano Capirchio
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
31
|
Walker M, Kuebler L, Goehring CM, Pichler BJ, Herfert K. Imaging SERT Availability in a Rat Model of L-DOPA-Induced Dyskinesia. Mol Imaging Biol 2021; 22:634-642. [PMID: 31392531 DOI: 10.1007/s11307-019-01418-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The development of L-DOPA-induced dyskinesia (LID) is one of the most severe side effects of chronic L-DOPA treatment in Parkinson's disease patients. [11C]DASB positron emission tomography (PET) provides a prominent tool to visualize and quantify serotonin transporter (SERT) pathology in vivo in patients and in animal models. To evaluate the effect of chronic L-DOPA treatment on SERT availability in an animal model of LID, we performed a longitudinal PET study. PROCEDURES Rats received a unilateral 6-hydroxydopamine (6-OHDA) lesion, and striatal and extrastriatal SERT expression levels were studied with [11C]DASB, a marker of SERT availability, before and after daily treatment with L-DOPA. Dyskinesias were evaluated at different time points over a period of 21 days. RESULTS [11C]DASB binding was found to be decreased after 6-OHDA lesions in the striatum, cortex, and hippocampus 5 weeks after 6-OHDA injection in the lesioned hemisphere of the rat brain. Chronic L-DOPA priming resulted in a relative preservation of SERT availability in the lesioned and healthy hemisphere compared to baseline measurements. CONCLUSIONS Our longitudinal PET data support a preservation of SERT availability after the induction of L-DOPA-induced dyskinesia, which is in line with previous reports in dyskinetic PD patients.
Collapse
Affiliation(s)
- Michael Walker
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Laura Kuebler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Chris Marc Goehring
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany.
| |
Collapse
|
32
|
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
|
33
|
Chesler K, Motz C, Vo H, Douglass A, Allen RS, Feola AJ, Pardue MT. Initiation of L-DOPA Treatment After Detection of Diabetes-Induced Retinal Dysfunction Reverses Retinopathy and Provides Neuroprotection in Rats. Transl Vis Sci Technol 2021; 10:8. [PMID: 34003986 PMCID: PMC8054623 DOI: 10.1167/tvst.10.4.8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose L-DOPA treatment initiated at the start of hyperglycemia preserves retinal and visual function in diabetic rats. Here, we investigated a more clinically relevant treatment strategy in which retinal and visual dysfunction designated the beginning of the therapeutic window for L-DOPA treatment. Methods Spatial frequency thresholds using optomotor response and oscillatory potential (OP) delays using electroretinograms were compared at baseline, 3, 6, and 10 weeks after streptozotocin (STZ) between diabetic and control rats. L-DOPA/carbidopa treatment (DOPA) or vehicle was delivered orally 5 days per week beginning at 3 weeks after STZ, when significant retinal and visual deficits were measured. At 10 weeks after STZ, retinas were collected to measure L-DOPA, dopamine, and 3,4-dihydroxyphenylacetic acid (DOPAC) levels using high-performance liquid chromatography. Results Spatial frequency thresholds decreased at 6 weeks in diabetic vehicle rats (28%), whereas diabetic DOPA rats had stable thresholds (<1%) that maintained to 10 weeks, creating significantly higher thresholds compared with diabetic vehicle rats (P < 0.0001). OP2 implicit times in response to dim, rod-driven stimuli were decreased in diabetic compared with control rats (3 weeks, P < 0.0001; 10 weeks, P < 0.01). With L-DOPA treatment, OP2 implicit times recovered in diabetic rats to be indistinguishable from control rats by 10 weeks after STZ. Rats treated with L-DOPA showed significantly increased retinal L-DOPA (P < 0.001) and dopamine levels (P < 0.05). Conclusions L-DOPA treatment started after the detection of retinal and visual dysfunction showed protective effects in diabetic rats. Translational Relevance Early retinal functional deficits induced by diabetes can be used to identify an earlier therapeutic window for L-DOPA treatment which protects from further vision loss and restores retinal function.
Collapse
Affiliation(s)
- Kyle Chesler
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Cara Motz
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Harrison Vo
- Atlanta VA Healthcare System, Atlanta, GA, USA
| | | | - Rachael S Allen
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew J Feola
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Atlanta VA Healthcare System, Atlanta, GA, USA.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
34
|
Song T, Li J, Mei S, Jia X, Yang H, Ye Y, Yuan J, Zhang Y, Lu J. Nigral Iron Deposition Is Associated With Levodopa-Induced Dyskinesia in Parkinson's Disease. Front Neurosci 2021; 15:647168. [PMID: 33828454 PMCID: PMC8019898 DOI: 10.3389/fnins.2021.647168] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/16/2021] [Indexed: 11/29/2022] Open
Abstract
Objective To investigate iron deposition in the substantia nigra (SN) of Parkinson’s disease (PD) patients associated with levodopa-induced dyskinesia (LID). Methods Seventeen PD patients with LID, 17 PD patients without LID, and 16 healthy controls were recruited for this study. The mean QSM values of the whole, left, and right SN were compared among the three groups. A multivariate logistic regression model was constructed to determine the factors associated with increased risk of LID. The receiver operating characteristic curve of the QSM value of SN in discriminating PD with and without LID was evaluated. Results The mean QSM values of the whole and right SN in the PD with LID were higher than those in the PD without LID (∗P = 0.03, ∗P = 0.03). Multivariate logistic regression analysis revealed that the QSM value of whole, left, or right SN was a predictor of the development of LID (∗P = 0.03, ∗P = 0.04, and ∗P = 0.04). The predictive accuracy of LID in adding the QSM value of the whole, left, and right SN to LID-related clinical risk factors was 70.6, 64.7, and 67.6%, respectively. The QSM cutoff values between PD with and without LID of the whole, left, and right SN were 148.3, 165.4, and 152.7 ppb, respectively. Conclusion This study provides the evidence of higher iron deposition in the SN of PD patients with LID than those without LID, suggesting that the QSM value of the SN may be a potential early diagnostic neuroimaging biomarker for LID.
Collapse
Affiliation(s)
- Tianbin Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Jiping Li
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shanshan Mei
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaofei Jia
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongwei Yang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yongquan Ye
- UIH America, Inc., Houston, TX, United States
| | - Jianmin Yuan
- Central Research Institute, UIH Group, Shanghai, China
| | - Yuqing Zhang
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| |
Collapse
|
35
|
De Deurwaerdère P, Chagraoui A, Di Giovanni G. Serotonin/dopamine interaction: Electrophysiological and neurochemical evidence. PROGRESS IN BRAIN RESEARCH 2021; 261:161-264. [PMID: 33785130 DOI: 10.1016/bs.pbr.2021.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interaction between serotonin (5-HT) and dopamine (DA) in the central nervous system (CNS) plays an important role in the adaptive properties of living animals to their environment. These are two modulatory, divergent systems shaping and regulating in a widespread manner the activity of neurobiological networks and their interaction. The concept of one interaction linking these two systems is rather elusive when looking at the mechanisms triggered by these two systems across the CNS. The great variety of their interacting mechanisms is in part due to the diversity of their neuronal origin, the density of their fibers in a given CNS region, the distinct expression of their numerous receptors in the CNS, the heterogeneity of their intracellular signaling pathway that depend on the cellular type expressing their receptors, and the state of activity of neurobiological networks, conditioning the outcome of their mutual influences. Thus, originally conceptualized as inhibition of 5-HT on DA neuron activity and DA neurotransmission, this interaction is nowadays considered as a multifaceted, mutual influence of these two systems in the regulation of CNS functions. These new ways of understanding this interaction are of utmost importance to envision the consequences of their dysfunctions underlined in several CNS diseases. It is also essential to conceive the mechanism of action of psychotropic drugs directly acting on their function including antipsychotic, antidepressant, antiparkinsonian, and drug of abuse together with the development of therapeutic strategies of Alzheimer's diseases, epilepsy, obsessional compulsive disorders. The 5-HT/DA interaction has a long history from the serendipitous discovery of antidepressants and antipsychotics to the future, rationalized treatments of CNS disorders.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Institut des Neurosciences Intégratives et Cognitives d'Aquitaine, UMR 5287, Bordeaux, France.
| | - Abdeslam Chagraoui
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), Normandie University, UNIROUEN, INSERM U1239, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
36
|
Serotonin/dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesia: An update. PROGRESS IN BRAIN RESEARCH 2021; 261:287-302. [PMID: 33785132 DOI: 10.1016/bs.pbr.2021.01.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ample evidence suggests that the serotonergic system plays a major role in several aspects of Parkinson's disease. In this review, we focus on the interplay between dopamine and serotonin in the appearance of L-DOPA-induced dyskinesia (LID), the most troublesome side effect of L-DOPA therapy. Indeed, while this drug exerts significant amelioration of motor symptoms during the first few years of treatment, eventually, most of patients experience dyskinesias, which limit the use of L-DOPA in advanced stages of disease. Here, we present the mechanisms underlying LID and the role of serotonin neurons, review preclinical and clinical data, and discuss possible therapeutic strategies.
Collapse
|
37
|
Wang Y, Yao L, Gao S, Zhang G, Zhang Q, Liu W, Zhou Y, Sun Y, Feng J, Liu J. Inhibition of striatal dopamine D 5 receptor attenuates levodopa-induced dyskinesia in a rat model of Parkinson's disease. Brain Res 2021; 1754:147266. [PMID: 33422541 DOI: 10.1016/j.brainres.2020.147266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/01/2020] [Accepted: 12/26/2020] [Indexed: 12/29/2022]
Abstract
Levodopa-induced dyskinesia (LID) is experienced by most patients of Parkinson's disease (PD) upon the long-term use of the dopamine precursor levodopa. Striatal dopaminergic signaling plays a critical role in the pathogenesis of LID through its interactions with dopamine receptors. The specific roles of striatal dopaminergic D5 receptors in the pathophysiological process of LID are still poorly established. In the study, we investigated the role of striatal dopamine D5 receptor in LID by using PD rats with or without dyskinetic symptoms after chronic levodopa administration. The experimental results showed that the expression level of D5 receptors in the sensorimotor striatum of dyskinetic rats is significantly higher than that of the non-dyskinetic controls. The administration of levodopa increased c-Fos expression in a subpopulation of sensorimotor striatum neurons of dyskinetic rats, but not in non-dyskinetic rats. The majority of the c-Fos+ neurons activated by levodopa in the striatum are positive for D5 receptor staining. Intrastriatal injection of D1-like (D1 and D5) dopamine receptor antagonist, SCH-23390, significantly inhibited dyskinetic behavior in dyskinetic rats after the injection of levodopa, meanwhile, intrastriatal administration of SKF-83959, a partial D5 receptor agonist, yielded significant dyskinetic movements in dyskinetic rats without levodopa. In contrast, intrastriatal perfusion of small interfering RNA directed against DRD5 downregulated D5 receptors expression and moderately inhibited dyskinetic behavior of dyskinetic animals. Our data suggested that the striatal dopamine D5 receptor might play a novel role in the pathophysiology of LID.
Collapse
Affiliation(s)
- Yong Wang
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China; Institute of Neuroscience, Xi'an Jiaotong University Health Science Center, Xi'an, PR China.
| | - Lu Yao
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China; Institute of Neuroscience, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Shasha Gao
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China; Institute of Neuroscience, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Gejuan Zhang
- Department of Neurology, Xi'an No. 3 Hospital, Xi'an 710018, PR China
| | - Qiongchi Zhang
- Undergraduate School, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Wanyuan Liu
- Undergraduate School, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yingqiong Zhou
- Undergraduate School, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yina Sun
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Jie Feng
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China; Institute of Neuroscience, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Jian Liu
- Deptartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China; Institute of Neuroscience, Xi'an Jiaotong University Health Science Center, Xi'an, PR China.
| |
Collapse
|
38
|
Serotonergic control of the glutamatergic neurons of the subthalamic nucleus. PROGRESS IN BRAIN RESEARCH 2021; 261:423-462. [PMID: 33785138 DOI: 10.1016/bs.pbr.2020.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The subthalamic nucleus (STN) houses a dense cluster of glutamatergic neurons that play a central role in the functional dynamics of the basal ganglia, a group of subcortical structures involved in the control of motor behaviors. Numerous anatomical, electrophysiological, neurochemical and behavioral studies have reported that serotonergic neurons from the midbrain raphe nuclei modulate the activity of STN neurons. Here, we describe this serotonergic innervation and the nature of the regulation exerted by serotonin (5-hydroxytryptamine, 5-HT) on STN neuron activity. This regulation can occur either directly within the STN or at distal sites, including other structures of the basal ganglia or cortex. The effect of 5-HT on STN neuronal activity involves several 5-HT receptor subtypes, including 5-HT1A, 5-HT1B, 5-HT2C and 5-HT4 receptors, which have garnered the highest attention on this topic. The multiple regulatory effects exerted by 5-HT are thought to be modified under pathological conditions, altering the activity of the STN, or due to the benefits and side effects of treatments used for Parkinson's disease, notably the dopamine precursor l-DOPA and high-frequency STN stimulation. Originally understood as a motor center, the STN is also associated with decision making and participates in mood regulation and cognitive performance, two domains of personality that are also regulated by 5-HT. The literature concerning the link between 5-HT and STN is already important, and the functional overlap is evident, but this link is still not entirely understood. The understanding of this link between 5-HT and STN should be increased due to the possible importance of this regulation in the control of fronto-STN loops and inherent motor and non-motor behaviors.
Collapse
|
39
|
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
|
40
|
Globus pallidus, but not entopeduncular nucleus, 6-OHDA-induced lesion attenuates L-Dopa-induced dyskinesia in the rat model of Parkinson's disease. Pharmacol Biochem Behav 2020; 197:173013. [DOI: 10.1016/j.pbb.2020.173013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
|
41
|
Vegas‐Suárez S, Pisanò CA, Requejo C, Bengoetxea H, Lafuente JV, Morari M, Miguelez C, Ugedo L. 6-Hydroxydopamine lesion and levodopa treatment modify the effect of buspirone in the substantia nigra pars reticulata. Br J Pharmacol 2020; 177:3957-3974. [PMID: 32464686 PMCID: PMC7429490 DOI: 10.1111/bph.15145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE l-DOPA-induced dyskinesia (LID) is considered a major complication in the treatment of Parkinson's disease (PD). Buspirone (5-HT1A partial agonist) have shown promising results in the treatment of PD and LID, however no 5-HT-based treatment has been approved in PD. The present study was aimed to investigate how the substantia nigra pars reticulata (SNr) is affected by buspirone and whether it is a good target to study 5-HT antidyskinetic treatments. EXPERIMENTAL APPROACH Buspirone was studied using in vivo single-unit, electrocorticogram, local field potential recordings along with microdialysis and immunohistochemistry in naïve/sham, 6-hydroxydopamine (6-OHDA)-lesioned or 6-OHDA-lesioned and l-DOPA-treated (6-OHDA/l-DOPA) rats. KEY RESULTS Local buspirone inhibited SNr neuron activity in all groups. However, systemic buspirone reduced burst activity in 6-OHDA-lesioned rats (with or without l-DOPA treatment), whereas 8-OH-DPAT, a full 5-HT1A agonist induced larger inhibitory effects in sham animals. Neither buspirone nor 8-OH-DPAT markedly modified the low-frequency oscillatory activity in the SNr or synchronization within the SNr with the cortex. In addition, local perfusion of buspirone increased GABA and glutamate release in the SNr of naïve and 6-OHDA-lesioned rats but no effect in 6-OHDA/l-DOPA rats. In the 6-OHDA/l-DOPA group, increased 5-HT transporter and decreased 5-HT1A receptor expression was found. CONCLUSIONS AND IMPLICATIONS The effects of buspirone in SNr are influenced by dopamine loss and l-DOPA treatment. The present results suggest that the regulation of burst activity of the SNr induced by DA loss may be a good target to test new drugs for the treatment of PD and LID.
Collapse
Affiliation(s)
- Sergio Vegas‐Suárez
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| | - Clarissa Anna Pisanò
- Department of Medical Sciences, Section of PharmacologyUniversity of FerraraFerraraItaly
- Neuroscience Center and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - Catalina Requejo
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Harkaitz Bengoetxea
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Jose Vicente Lafuente
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Michele Morari
- Department of Medical Sciences, Section of PharmacologyUniversity of FerraraFerraraItaly
- Neuroscience Center and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| |
Collapse
|
42
|
Calabrese V, Di Maio A, Marino G, Cardinale A, Natale G, De Rosa A, Campanelli F, Mancini M, Napolitano F, Avallone L, Calabresi P, Usiello A, Ghiglieri V, Picconi B. Rapamycin, by Inhibiting mTORC1 Signaling, Prevents the Loss of Striatal Bidirectional Synaptic Plasticity in a Rat Model of L-DOPA-Induced Dyskinesia. Front Aging Neurosci 2020; 12:230. [PMID: 32848709 PMCID: PMC7431470 DOI: 10.3389/fnagi.2020.00230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
Levodopa (L-DOPA) treatment is the main gold-standard therapy for Parkinson disease (PD). Besides good antiparkinsonian effects, prolonged use of this drug is associated to the development of involuntary movements known as L-DOPA-induced dyskinesia (LID). L-DOPA-induced dyskinesia is linked to a sensitization of dopamine (DA) D1 receptors located on spiny projection neurons (SPNs) of the dorsal striatum. Several evidences have shown that the emergence of LID can be related to striatal D1/cAMP/PKA/DARPP-32 and extracellular signal-regulated kinases (ERK1/2) pathway overactivation associated to aberrant N-methyl-d-aspartate (NMDA) receptor function. In addition, within striatum, ERK1/2 is also able to modulate in a D1 receptor-dependent manner the activity of the mammalian target of rapamycin complex 1 (mTORC1) pathway under DA depletion and L-DOPA therapy. Consistently, increased mTORC1 signaling appears during chronic administration of L-DOPA and shows a high correlation with the severity of dyskinesia. Furthermore, the abnormal activation of the D1/PKA/DARPP-32 cascade is paralleled by increased phosphorylation of the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor at the PKA Ser845 site. The GluA1 promotes excitatory AMPA receptor-mediated transmission and may be implicated in the alterations found at the corticostriatal synapses of dyskinetic animals. In our study, we investigated the role of mTORC1 pathway activation in modulating bidirectional striatal synaptic plasticity in L-DOPA-treated parkinsonian rats. Inhibition of mTORC1 by coadministration of rapamycin to L-DOPA was able to limit the magnitude of LID expression, accounting for a therapeutic effect of this drug. In particular, behavioral data showed that, in L-DOPA-treated rats, rapamycin administration induced a selective decrease of distinct components of abnormal involuntary movements (i.e., axial and orolingual dyskinesia). Furthermore, ex vivo patch clamp and intracellular recordings of SPNs revealed that pharmacological inhibition of mTORC1 also resulted associated with a physiological bidirectional plasticity, when compared to dyskinetic rats treated with L-DOPA alone. This study uncovers the important role of mTORC1 inhibition to prevent the loss of striatal bidirectional plasticity under chronic L-DOPA treatment in rodent models of PD.
Collapse
Affiliation(s)
- Valeria Calabrese
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Gioia Marino
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Antonella Cardinale
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | - Giuseppina Natale
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Arianna De Rosa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Campanelli
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Mancini
- Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesco Napolitano
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Paolo Calabresi
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DISTABIF), University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Veronica Ghiglieri
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Università Telematica San Raffaele, Rome, Italy
| |
Collapse
|
43
|
AlShimemeri S, Fox SH, Visanji NP. Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update. Expert Opin Emerg Drugs 2020; 25:131-144. [DOI: 10.1080/14728214.2020.1763954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sohaila AlShimemeri
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Susan H Fox
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
44
|
Meloni M, Puligheddu M, Sanna F, Cannas A, Farris R, Tronci E, Figorilli M, Defazio G, Carta M. Efficacy and safety of 5-Hydroxytryptophan on levodopa-induced motor complications in Parkinson's disease: A preliminary finding. J Neurol Sci 2020; 415:116869. [PMID: 32464351 DOI: 10.1016/j.jns.2020.116869] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/02/2020] [Accepted: 04/24/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND AND PURPOSE Several studies have indicated that altered serotonergic neurotransmission may contribute to the motor features commonly associated with Parkinson's disease (PD) drug treatment such as levodopa-induced dyskinesias (LIDs). 5-Hydroxytryptophan (5-HTP) is the immediate precursor of serotonin. We have recently demonstrated that 5-HTP produces significant antidyskinetic effects in a rat model of PD. To date, there has been inconsistent research on the use of 5-HTP in PD. The purpose of this study was to compare the effects of 5-HTP versus placebo on levodopa-induced motor complications in PD patients. MATERIAL AND METHODS A single-center, randomized, double-blind placebo-controlled cross-over study was performed. A total of 12 PD patients were diagnosed with LIDs and motor fluctuactions and subsequently were randomized to intervention; 11 subjects completed the entire 16-week protocol. Patients received placebo or 50 mg of 5-HTP daily in a cross-over design over a period of 4 weeks. For the assessment of efficacy on the motor functions and motor complications, the UPDRS (parts III and IV), Unified Dyskinesia Rating Scale (UDysRS), Wearing-Off Questionnaire (WOQ-19) and the self-reported 24-h home dyskinesia diaries were obtained at baseline and weeks 4, 8, 12 and 16 (T-end). RESULTS Repeated measures analysis revealed a significant improvement of LIDs during the 50 mg 5-HTP treatment as assessed by the UDysRS and UPDRS-IV scores. CONCLUSIONS This study provides preliminary evidence of clinical benefit of 5-HTP against LIDs in PD. Larger studies with a longer treatment duration and a wider range of doses are warranted to corroborate these findings.
Collapse
Affiliation(s)
- Mario Meloni
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.
| | - Monica Puligheddu
- Sleep Disorders Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Medical Sciences and Public Health, Neurology Unit, University of Cagliari and AOU Cagliari, Monserrato, Cagliari, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonino Cannas
- Department of Medical Sciences and Public Health, Neurology Unit, University of Cagliari and AOU Cagliari, Monserrato, Cagliari, Italy
| | - Rita Farris
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Michela Figorilli
- Sleep Disorders Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Giovanni Defazio
- Department of Medical Sciences and Public Health, Neurology Unit, University of Cagliari and AOU Cagliari, Monserrato, Cagliari, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
45
|
Sugiyama E, Skelly AN, Suematsu M, Sugiura Y. In situ imaging of monoamine localization and dynamics. Pharmacol Ther 2020; 208:107478. [DOI: 10.1016/j.pharmthera.2020.107478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 11/22/2019] [Indexed: 01/06/2023]
|
46
|
Lowes H, Pyle A, Santibanez-Koref M, Hudson G. Circulating cell-free mitochondrial DNA levels in Parkinson's disease are influenced by treatment. Mol Neurodegener 2020; 15:10. [PMID: 32070373 PMCID: PMC7029508 DOI: 10.1186/s13024-020-00362-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Several studies have linked circulating cell-free mitochondrial DNA (ccf-mtDNA) to human disease. In particular, reduced ccf-mtDNA levels in the cerebrospinal fluid (CSF) of both Alzheimer's and Parkinson's disease (PD) patients have raised the hypothesis that ccf-mtDNA could be used as a biomarker for neurodegenerative disease onset and progression. However, how a reduction of CSF ccf-mtDNA levels relates to neurodegeneration remains unclear. Many factors are likely to influence ccf-mtDNA levels, such as concomitant therapeutic treatment and comorbidities. In this study we aimed to investigate these factors, quantifying CSF ccf-mtDNA from the Parkinson's Progression Markers Initiative in 372 PD patients and 159 matched controls at two time points. We found that ccf-mtDNA levels appear significantly reduced in PD cases when compared to matched controls and are associated with cognitive impairment. However, our data indicate that this reduction in ccf-mtDNA is also associated with the commencement, type and duration of treatment. Additionally, we found that ccf-mtDNA levels are associated with comorbidities such as depression and insomnia, however this was only significant if measured in the absence of treatment. We conclude that in PD, similar to reports in HIV and sepsis, comorbidities and treatment can both influence ccf-mtDNA homeostasis, raising the possibility that ccf-mtDNA may be useful as a biomarker for treatment response or the development of secondary phenotypes. Given that, clinically, PD manifests often decades after neurodegeneration begins, predicting who will develop disease is important. Also, identifying patients who will respond to existing treatments or develop secondary phenotypes will have increased clinical importance as PD incidence rises.
Collapse
Affiliation(s)
- Hannah Lowes
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ UK
| | - Angela Pyle
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH UK
| | | | - Gavin Hudson
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ UK
| |
Collapse
|
47
|
Chagraoui A, Boulain M, Juvin L, Anouar Y, Barrière G, De Deurwaerdère P. L-DOPA in Parkinson's Disease: Looking at the "False" Neurotransmitters and Their Meaning. Int J Mol Sci 2019; 21:ijms21010294. [PMID: 31906250 PMCID: PMC6981630 DOI: 10.3390/ijms21010294] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) has been successfully used in the treatment of Parkinson’s disease (PD) for more than 50 years. It fulfilled the criteria to cross the blood–brain barrier and counteract the biochemical defect of dopamine (DA). It remarkably worked after some adjustments in line with the initial hypothesis, leaving a poor place to the plethora of mechanisms involving other neurotransmitters or mechanisms of action beyond newly synthesized DA itself. Yet, its mechanism of action is far from clear. It involves numerous distinct cell populations and does not mimic the mechanism of action of dopaminergic agonists. L-DOPA-derived DA is mainly released by serotonergic neurons as a false neurotransmitter, and serotonergic neurons are involved in L-DOPA-induced dyskinesia. The brain pattern and magnitude of DA extracellular levels together with this status of false neurotransmitters suggest that the striatal effects of DA via this mechanism would be minimal. Other metabolic products coming from newly formed DA or through the metabolism of L-DOPA itself could be involved. These compounds can be trace amines and derivatives. They could accumulate within the terminals of the remaining monoaminergic neurons. These “false neurotransmitters,” also known for some of them as inducing an “amphetamine-like” mechanism, could reduce the content of biogenic amines in terminals of monoaminergic neurons, thereby impairing the exocytotic process of monoamines including L-DOPA-induced DA extracellular outflow. The aim of this review is to present the mechanism of action of L-DOPA with a specific attention to “false neurotransmission.”
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), Normandie University, UNIROUEN, INSERM, U1239 CHU de Rouen, 76000 Rouen, France; (A.C.); (Y.A.)
- Department of Medical Biochemistry, Rouen University Hospital, CHU de Rouen, 76000 Rouen, France
| | - Marie Boulain
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 33076 Bordeaux CEDEX, France; (M.B.); (L.J.); (G.B.)
| | - Laurent Juvin
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 33076 Bordeaux CEDEX, France; (M.B.); (L.J.); (G.B.)
| | - Youssef Anouar
- Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), Normandie University, UNIROUEN, INSERM, U1239 CHU de Rouen, 76000 Rouen, France; (A.C.); (Y.A.)
| | - Grégory Barrière
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 33076 Bordeaux CEDEX, France; (M.B.); (L.J.); (G.B.)
| | - Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 33076 Bordeaux CEDEX, France; (M.B.); (L.J.); (G.B.)
- Correspondence: ; Tel.: +33-0-557-57-12-90
| |
Collapse
|
48
|
Aristieta A, Ruiz-Ortega J, Morera-Herreras T, Miguelez C, Ugedo L. Acute L-DOPA administration reverses changes in firing pattern and low frequency oscillatory activity in the entopeduncular nucleus from long term L-DOPA treated 6-OHDA-lesioned rats. Exp Neurol 2019; 322:113036. [DOI: 10.1016/j.expneurol.2019.113036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023]
|
49
|
Iyer V, Vo Q, Mell A, Chinniah S, Zenerovitz A, Venkiteswaran K, Kunselman AR, Fang J, Subramanian T. Acute levodopa dosing around-the-clock ameliorates REM sleep without atonia in hemiparkinsonian rats. NPJ PARKINSONS DISEASE 2019; 5:27. [PMID: 31815176 PMCID: PMC6884572 DOI: 10.1038/s41531-019-0096-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/21/2019] [Indexed: 01/08/2023]
Abstract
Rapid-eye-movement (REM) sleep without atonia (RSWA), a marker of REM sleep behavior disorder (RBD), is frequently comorbid with Parkinson's disease (PD). Although rodent models are commonly used for studying PD, the neurobiological and behavioral correlates of RBD remain poorly understood. Therefore, we developed a behavior-based criteria to identify RSWA in the hemiparkinsonian rat model of PD. Video recordings of rats were analyzed, to develop a criteria consisting of behavioral signs that occurred during polysomnographically confirmed epochs of sleep-wake stages. The sleep-slouch, a postural shift of the body or head caused only by gravity, was identified as a unique behavioral sign of REM sleep onset and was altered in hemiparkinsonian rats during RSWA. There was a significant correlation between the behavior-based criteria and polysomnograms for all sleep-wake stages in control but not hemiparkinsonian rats indicating a deterioration of sleep-wake architecture in parkinsonism. We then tested the efficacy of levodopa in ameliorating RSWA using intermittent and around-the-clock (ATC) dosing regimens. ATC levodopa dosing at 4 mg/kg for 48 h caused a significant reduction of RSWA as measured by polysomnography and the behavioral-based criteria along with an amelioration of forelimb motor deficits. Our findings show that the phenomenological correlates of RSWA can be reliably characterized in the hemiparkinsonian rat model. ATC levodopa administration ameliorates RSWA in this model without deleterious consequences to the overall sleep-wake architecture and therapeutic benefits for parkinsonian motor deficits. These findings suggest that further study may allow for the application of a similar approach to treat RBD in PD patients.
Collapse
Affiliation(s)
- Vishakh Iyer
- 1Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN USA
| | - Quynh Vo
- 2Department of Neurology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Anthony Mell
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Siven Chinniah
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Ashley Zenerovitz
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Kala Venkiteswaran
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Allen R Kunselman
- 4Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Jidong Fang
- 5Department of Psychiatry, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Thyagarajan Subramanian
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| |
Collapse
|
50
|
Sugiyama E, Guerrini MM, Honda K, Hattori Y, Abe M, Källback P, Andrén PE, Tanaka KF, Setou M, Fagarasan S, Suematsu M, Sugiura Y. Detection of a High-Turnover Serotonin Circuit in the Mouse Brain Using Mass Spectrometry Imaging. iScience 2019; 20:359-372. [PMID: 31614319 PMCID: PMC6818351 DOI: 10.1016/j.isci.2019.09.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 07/12/2019] [Accepted: 09/24/2019] [Indexed: 12/31/2022] Open
Abstract
Monoamine neurotransmitters are released by specialized neurons regulating behavioral, motor, and cognitive functions. Although the localization of monoaminergic neurons in the brain is well known, the distribution and kinetics of monoamines remain unclear. Here, we generated a murine brain atlas of serotonin (5-HT), dopamine (DA), and norepinephrine (NE) levels using mass spectrometry imaging (MSI). We found several nuclei rich in both 5-HT and a catecholamine (DA or NE) and identified the paraventricular nucleus of the thalamus (PVT), where 5-HT and NE are co-localized. The analysis of 5-HT fluctuations in response to acute tryptophan depletion and infusion of isotope-labeled tryptophan in vivo revealed a close kinetic association between the raphe nuclei, PVT, and amygdala but not the other nuclei. Our findings imply the existence of a highly dynamic 5-HT-mediated raphe to PVT pathway that likely plays a role in the brain monoamine system. A murine brain atlas of monoamine (5-HT, DA, NE) levels was generated via MS imaging We identified several nuclei rich in both 5-HT and a catecholamine (DA or NE) The paraventricular nucleus of the thalamus (PVT) had high levels of 5-HT and NE The level of 5-HT in raphe to PVT pathway changed dynamically in response to blood Trp level
Collapse
Affiliation(s)
- Eiji Sugiyama
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Matteo M Guerrini
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Tsurumi Ward, Suehirocho, 1 Chome-7-22, Yokohama, Kanagawa Prefecture 230-0045, Japan
| | - Kurara Honda
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yuko Hattori
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| | - Patrik Källback
- Medical Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591 BMC, 75124 Uppsala, Sweden; Science for Life Laboratory, National Resource for Mass Spectrometry Imaging, Uppsala University, Box 591 BMC, 75124 Uppsala, Sweden
| | - Per E Andrén
- Medical Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591 BMC, 75124 Uppsala, Sweden; Science for Life Laboratory, National Resource for Mass Spectrometry Imaging, Uppsala University, Box 591 BMC, 75124 Uppsala, Sweden
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy and International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Tsurumi Ward, Suehirocho, 1 Chome-7-22, Yokohama, Kanagawa Prefecture 230-0045, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| |
Collapse
|