1
|
Guimarães RP, de Resende MCS, Tavares MM, Belardinelli de Azevedo C, Ruiz MCM, Mortari MR. Construct, Face, and Predictive Validity of Parkinson's Disease Rodent Models. Int J Mol Sci 2024; 25:8971. [PMID: 39201659 PMCID: PMC11354451 DOI: 10.3390/ijms25168971] [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: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally. Current drugs only alleviate symptoms without halting disease progression, making rodent models essential for researching new therapies and understanding the disease better. However, selecting the right model is challenging due to the numerous models and protocols available. Key factors in model selection include construct, face, and predictive validity. Construct validity ensures the model replicates pathological changes seen in human PD, focusing on dopaminergic neurodegeneration and a-synuclein aggregation. Face validity ensures the model's symptoms mirror those in humans, primarily reproducing motor and non-motor symptoms. Predictive validity assesses if treatment responses in animals will reflect those in humans, typically involving classical pharmacotherapies and surgical procedures. This review highlights the primary characteristics of PD and how these characteristics are validated experimentally according to the three criteria. Additionally, it serves as a valuable tool for researchers in selecting the most appropriate animal model based on established validation criteria.
Collapse
Affiliation(s)
- Rayanne Poletti Guimarães
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Maria Clara Souza de Resende
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Mesquita Tavares
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Caio Belardinelli de Azevedo
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Cesar Merino Ruiz
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
- Neurological Rehabilitation Unit, Sarah Network of Rehabilitation Hospitals, Brasília 70335-901, Brazil
| | - Márcia Renata Mortari
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| |
Collapse
|
2
|
Cesaroni V, Blandini F, Cerri S. Dyskinesia and Parkinson's disease: animal model, drug targets, and agents in preclinical testing. Expert Opin Ther Targets 2022; 26:837-851. [PMID: 36469635 DOI: 10.1080/14728222.2022.2153036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. PD patients exhibit a classic spectrum of motor symptoms, arising when dopamine neurons in the substantia nigra pars compacta are reduced by 60%. The dopamine precursor L-DOPA represents the most effective therapy for improving PD motor dysfunctions, thus far available. Unfortunately, long-term treatment with L-DOPA is associated with the development of severe side effects, resulting in abnormal involuntary movements termed levodopa-induced dyskinesia (LID). Amantadine is the only drug currently approved for the treatment of LID indicating that LID management is still an unmet need in PD and encouraging the search for novel anti-dyskinetic drugs or the assessment of combined therapies with different molecular targets. AREAS COVERED This review provides an overview of the main preclinical models used to study LID and of the latest preclinical evidence on experimental and clinically available pharmacological approaches targeting non-dopaminergic systems. EXPERT OPINION LIDs are supported by complex molecular and neurobiological mechanisms that are still being studied today. This complexity suggests the need of developing personalized pharmacological approach to obtain an effective amelioration of LID condition and improve the quality of life of PD patients.
Collapse
Affiliation(s)
- Valentina Cesaroni
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation 27100, Pavia, Italy
| | - Fabio Blandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico 20122, Milan, Italy
| | - Silvia Cerri
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation 27100, Pavia, Italy
| |
Collapse
|
3
|
Synergistic effect of serotonin 1A and serotonin 1B/D receptor agonists in the treatment of L-DOPA-induced dyskinesia in 6-hydroxydopamine-lesioned rats. Exp Neurol 2022; 358:114209. [PMID: 35988699 DOI: 10.1016/j.expneurol.2022.114209] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The gold standard for symptomatic relief of Parkinson's disease (PD) is L-DOPA. However, long-term treatment often leads to motor complications such as L-DOPA-induced dyskinesia (LID). While amantadine (Gocovri™) is the only approved therapy for dyskinesia in PD patients on the American market, it is associated with neurological side effects and limited efficacy. Thus, there remains a high unmet need for addressing LID in PD patients worldwide. OBJECTIVE The objective of this study was to evaluate the efficacy, safety and performance compared to approved treatments of the serotonin receptor 1A (5-HT1A) and 5-HT1B/D agonists buspirone and zolmitriptan in the 6-hydroxydopamine unilaterally lesioned rat model for PD. METHODS The hemiparkinsonian 6-OHDA-lesioned rats underwent chronic treatment with L-DOPA to induce dyskinesia and were subsequently used for efficacy testing of buspirone, zolmitriptan and comparison with amantadine, measured as abnormal involuntary movement (AIM) scores after L-DOPA challenge. Safety testing was performed in model and naïve animals using forelimb adjusting, rotarod and open field tests. RESULTS 5-HT1A and 5-HT1B/D agonism effectively reduced AIM scores in a synergistic manner. The drug combination of buspirone and zolmitriptan was safe and did not lead to tolerance development following sub-chronic administration. Head-to-head comparison with amantadine showed superior performance of buspirone and zolmitriptan in the model. CONCLUSIONS The strong anti-dyskinetic effect found with combined 5-HT1A and 5-HT1B/D agonism renders buspirone and zolmitriptan together a meaningful treatment for LID in PD.
Collapse
|
4
|
Hansen CA, Miller DR, Annarumma S, Rusch CT, Ramirez-Zamora A, Khoshbouei H. Levodopa-induced dyskinesia: a historical review of Parkinson's disease, dopamine, and modern advancements in research and treatment. J Neurol 2022; 269:2892-2909. [PMID: 35039902 DOI: 10.1007/s00415-022-10963-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/01/2022]
Abstract
Over the past two decades, animal models of Parkinson's disease (PD) have helped to determine the plausible underlying mechanism of levo-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia following L-DOPA treatment. However, our understanding of the mechanisms related to this phenomenon remains incomplete. The purpose of this manuscript is to provide a comprehensive review of treatment protocols used for assessing the occurrence of L-DOPA-induced dyskinesia, L-DOPA absorption, distribution, drug/food interaction, and discuss current strategies and future directions. This review offers a historical perspective using L-DOPA in animal models of PD and the occurrence of L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Carissa A Hansen
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Douglas R Miller
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Stephanie Annarumma
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Carley T Rusch
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA.,Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Adolfo Ramirez-Zamora
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Fu XX, Wang J, Cai HY, Jiang H, Jiang JZ, Chen HH, Han S. Co-Application of C16 and Ang-1 Improves the Effects of Levodopa in Parkinson Disease Treatment. J Inflamm Res 2022; 15:3797-3814. [PMID: 35836722 PMCID: PMC9273834 DOI: 10.2147/jir.s368291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Levodopa is regarded as a standard medication in Parkinson disease (PD) treatment. However, long-term administration of levodopa leads to levodopa-induced dyskinesia (LID), which can markedly affect patient quality of life. Previous studies have shown that neuroinflammation in the brain plays a role in LID and increases potential neuroinflammatory mediators associated with the side effects of levodopa. OBJECTIVE The treatment effect of C16 (a peptide that competitively binds integrin αvβ3 and inhibits inflammatory cell infiltration) and angiopoietin-1 (Ang-1; a vascular endothelial growth factor vital for blood vessel protection), along with levodopa, was evaluated in a rodent model of PD. METHODS We administered a combination of C16 and Ang-1 in a rodent model of PD induced by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). Seventy-five mice were randomly divided into five treatment groups: control, vehicle, levodopa, C16+Ang-1, and levodopa+C16+Ang-1. Behavioral, histological, and electrophysiological experiments were used to determine neuron function and recovery. RESULTS The results showed that C16+Ang-1 treatment alleviated neuroinflammation in the CNS and promoted the recovery effects of levodopa on neural function. CONCLUSION Our study suggests that C16+Ang-1 can compensate for the shortcomings of levodopa, improve the CNS microenvironment, and ameliorate the effects of levodopa. This treatment strategy could be developed as a combinatorial therapeutic in the future.
Collapse
Affiliation(s)
- Xiao-Xiao Fu
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jin Wang
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hua-Ying Cai
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hong Jiang
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jin-Zhan Jiang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, People’s Republic of China
| | - Hao-Hao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, People’s Republic of China
- Hao-Hao Chen, Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, 1188 Wuzhou Steet, Jinhua, People’s Republic of China, Tel +86-579-82265128, Fax +86-579-82265110, Email
| | - Shu Han
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
- Correspondence: Shu Han, Institute of Anatomy, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, 866 Yuhangtang Road, Hangzhou, People’s Republic of China, Tel +86-571-88208160, Fax +86-571-88208094, Email
| |
Collapse
|
6
|
Kreis SL, Luhmann HJ, Ciolac D, Groppa S, Muthuraman M. Translational Model of Cortical Premotor-Motor Networks. Cereb Cortex 2021; 32:2621-2634. [PMID: 34689188 PMCID: PMC9201593 DOI: 10.1093/cercor/bhab369] [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: 07/19/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Deciphering the physiological patterns of motor network connectivity is a prerequisite to elucidate aberrant oscillatory transformations and elaborate robust translational models of movement disorders. In the proposed translational approach, we studied the connectivity between premotor (PMC) and primary motor cortex (M1) by recording high-density electroencephalography in humans and between caudal (CFA) and rostral forelimb (RFA) areas by recording multi-site extracellular activity in mice to obtain spectral power, functional and effective connectivity. We identified a significantly higher spectral power in β- and γ-bands in M1compared to PMC and similarly in mice CFA layers (L) 2/3 and 5 compared to RFA. We found a strong functional β-band connectivity between PMC and M1 in humans and between CFA L6 and RFA L5 in mice. We observed that in both humans and mice the direction of information flow mediated by β- and γ-band oscillations was predominantly from PMC toward M1 and from RFA to CFA, respectively. Combining spectral power, functional and effective connectivity, we revealed clear similarities between human PMC-M1 connections and mice RFA-CFA network. We propose that reciprocal connectivity of mice RFA-CFA circuitry presents a suitable model for analysis of motor control and physiological PMC-M1 functioning or pathological transformations within this network.
Collapse
Affiliation(s)
- Svenja L Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55128, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55128, Germany
| | - Dumitru Ciolac
- Section of Movement Disorders and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55131, Germany.,Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau MD-2001, Republic of Moldova
| | - Sergiu Groppa
- Section of Movement Disorders and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55131, Germany
| | - Muthuraman Muthuraman
- Section of Movement Disorders and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55131, Germany
| |
Collapse
|
7
|
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
|
8
|
Can adenosine A 2A receptor antagonists modify motor behavior and dyskinesia in experimental models of Parkinson's disease? Parkinsonism Relat Disord 2020; 80 Suppl 1:S21-S27. [PMID: 33349576 DOI: 10.1016/j.parkreldis.2020.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 11/24/2022]
Abstract
Current treatment of the motor symptoms of Parkinson's disease (PD) focuses on dopamine replacement therapies. While these treatments are initially highly effective, with long-term use and disease progression, the therapeutic response is often limited by the development of motor complications, dopaminergic side effects, and residual unresponsive motor and non-motor symptoms. An alternative or additive treatment approach may be to target non-dopaminergic receptors within the motor control pathways, which function to modulate basal ganglia output. Adenosine A2A receptors are one potential non-dopaminergic target as they are selectively localized to the basal ganglia and to the indirect output pathway known to modulate the striato-thalamo-cortical loops critical to the expression of the motor symptoms of PD. This paper reviews the preclinical evidence base for the ability of adenosine A2A receptor blockade to influence motor function and modulate dyskinesia expression. There is consensus that adenosine A2A receptor antagonists - administered either as a monotherapy or in combination with l-DOPA or dopamine agonists - improve motor function in both rodent and primate models of PD, and should be effective for treating the motor symptoms of PD in humans. Importantly, the improvements in motor function were seen in the absence of dyskinesia. The introduction of a non-dopaminergic approach to modifying basal ganglia function provides a useful addition to the range of available therapies for treating PD, and there is a rational basis for a drug that focuses on modifying basal ganglia output.
Collapse
|
9
|
Blosser JA, Podolsky E, Lee D. L-DOPA-Induced Dyskinesia in a Genetic Drosophila Model of Parkinson's Disease. Exp Neurobiol 2020; 29:273-284. [PMID: 32921640 PMCID: PMC7492844 DOI: 10.5607/en20028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 01/05/2023] Open
Abstract
Motor symptoms in Parkinson’s disease (PD) are directly related to the reduction of a neurotransmitter dopamine. Therefore, its precursor L-DOPA became the gold standard for PD treatment. However, chronic use of L-DOPA causes uncontrollable, involuntary movements, called L-DOPA-induced dyskinesia (LID) in the majority of PD patients. LID is complicated and very difficult to manage. Current rodent and non-human primate models have been developed to study LID mainly using neurotoxins. Therefore, it is necessary to develop a LID animal model with defects in genetic factors causing PD in order to study the relation between LID and PD genes such as α-synuclein. In this study, we first showed that a low concentration of L-DOPA (100 µM) rescues locomotion defects (i.e., speed, angular velocity, pause time) in Drosophila larvae expressing human mutant α-synuclein (A53T). This A53T larval model of PD was used to further examine dyskinetic behaviors. High concentrations of L-DOPA (5 or 10 mM) causes hyperactivity such as body bending behavior (BBB) in A53T larva, which resembles axial dyskinesia in rodents. Using ImageJ plugins and other third party software, dyskinetic BBB has been accurately and efficiently quantified. Further, we showed that a dopamine agonist pramipexole (PRX) partially rescues BBB caused by high L-DOPA. Our Drosophila genetic LID model will provide an important experimental platform to examine molecular and cellular mechanisms underlying LID, to study the role of PD causing genes in the development of LID, and to identify potential targets to slow/reverse LID pathology.
Collapse
Affiliation(s)
- Joshua A Blosser
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Eric Podolsky
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
10
|
Espadas I, Keifman E, Palomo-Garo C, Burgaz S, García C, Fernández-Ruiz J, Moratalla R. Beneficial effects of the phytocannabinoid Δ 9-THCV in L-DOPA-induced dyskinesia in Parkinson's disease. Neurobiol Dis 2020; 141:104892. [PMID: 32387338 DOI: 10.1016/j.nbd.2020.104892] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
The antioxidant and CB2 receptor agonist properties of Δ9-tetrahydrocannabivarin (Δ9-THCV) afforded neuroprotection in experimental Parkinson's disease (PD), whereas its CB1 receptor antagonist profile at doses lower than 5 mg/kg caused anti-hypokinetic effects. In the present study, we investigated the anti-dyskinetic potential of Δ9-THCV (administered i.p. at 2 mg/kg for two weeks), which had not been investigated before. This objective was investigated after inducing dyskinesia by repeated administration of L-DOPA (i.p. at 10 mg/kg) in a genetic model of dopaminergic deficiency, Pitx3ak mutant mice, which serves as a useful model for testing anti-dyskinetic agents. The daily treatment of these mice with L-DOPA for two weeks progressively increased the time spent in abnormal involuntary movements (AIMs) and elevated their horizontal and vertical activities (as measured in a computer-aided actimeter), signs that reflected the dyskinetic state of these mice. Interestingly, when combined with L-DOPA from the first injection, Δ9-THCV delayed the appearance of all these signs and decreased their intensity, with a reduction in the levels of FosB protein and the histone pAcH3 (measured by immunohistochemistry), which had previously been found to be elevated in the basal ganglia in L-DOPA-induced dyskinesia. In addition to the anti-dyskinetic effects of Δ9-THCV when administered at the onset of L-DOPA treatment, Δ9-THCV was also effective in attenuating the intensity of dyskinesia when administered for three consecutive days once these signs were already present (two weeks after the onset of L-DOPA treatment). In summary, our data support the anti-dyskinetic potential of Δ9-THCV, both to delay the occurrence and to attenuate the magnitude of dyskinetic signs. Although further studies are clearly required to determine the clinical significance of these data in humans, the results nevertheless situate Δ9-THCV in a promising position for developing a cannabinoid-based therapy for patients with PD.
Collapse
Affiliation(s)
- Isabel Espadas
- Instituto Cajal-CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | | | - Cristina Palomo-Garo
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Sonia Burgaz
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Concepción García
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernández-Ruiz
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Rosario Moratalla
- Instituto Cajal-CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain.
| |
Collapse
|
11
|
Waters S, Sonesson C, Svensson P, Tedroff J, Carta M, Ljung E, Gunnergren J, Edling M, Svanberg B, Fagerberg A, Kullingsjö J, Hjorth S, Waters N. Preclinical Pharmacology of [2-(3-Fluoro-5-Methanesulfonyl-phenoxy)Ethyl](Propyl)amine (IRL790), a Novel Dopamine Transmission Modulator for the Treatment of Motor and Psychiatric Complications in Parkinson Disease. J Pharmacol Exp Ther 2020; 374:113-125. [PMID: 32358046 DOI: 10.1124/jpet.119.264226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/02/2020] [Indexed: 12/23/2022] Open
Abstract
IRL790 ([2-(3-fluoro-5-methanesulfonylphenoxy)ethyl](propyl)amine, mesdopetam) is a novel compound in development for the clinical management of motor and psychiatric disabilities in Parkinson disease. The discovery of IRL790 was made applying a systems pharmacology approach based on in vivo response profiling. The chemical design idea was to develop a new type of DA D3/D2 receptor type antagonist built on agonist rather than antagonist structural motifs. We hypothesized that such a dopamine antagonist with physicochemical properties similar to agonists would exert antidyskinetic and antipsychotic effects in states of dysregulated dopaminergic signaling while having little negative impact on physiologic dopamine transmission and, hence, minimal liability for side effects related to dopamine-dependent functions. At the level of in vivo pharmacology, IRL790 displays balancing effects on aberrant motor phenotypes, reducing l-DOPA-induced dyskinesias in the rodent 6-hydroxydopamine lesion model and reducing psychostimulant-induced locomotor hyperactivity elicited by pretreatment with either d-amphetamine or dizocilpine, without negatively impacting normal motor performance. Thus, IRL790 has the ability to normalize the behavioral phenotype in hyperdopaminergic as well as hypoglutamatergic states. Neurochemical and immediate early gene (IEG) response profiles suggest modulation of DA neurotransmission, with some features, such as increased DA metabolites and extracellular DA, shared by atypical antipsychotics and others, such as increased frontal cortex IEGs, unique to IRL790. IRL790 also increases extracellular levels of acetylcholine in the prefrontal cortex and ventral hippocampus. At the receptor level, IRL790 appears to act as a preferential DA D3 receptor antagonist. Computational docking studies support preferential affinity at D3 receptors with an agonist-like binding mode. SIGNIFICANCE STATEMENT: This paper reports preclinical pharmacology along with molecular modeling results on IRL790, a novel compound in clinical development for the treatment of motor and psychiatric complications in advanced Parkinson disease. IRL790 is active in models of perturbed dopaminergic and glutamatergic signaling, including rodent 6-hydroxydopamine l-DOPA-induced dyskinesias and psychostimulant-induced hyperactivity, in a dose range that does not impair normal behavior. This effect profile is attributed to interactions at dopamine D2/D3 receptors, with a 6- to 8-fold preference for the D3 subtype.
Collapse
Affiliation(s)
- Susanna Waters
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Clas Sonesson
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Peder Svensson
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Joakim Tedroff
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Manolo Carta
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Elisabeth Ljung
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Jenny Gunnergren
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Malin Edling
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Boel Svanberg
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Anne Fagerberg
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Johan Kullingsjö
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Stephan Hjorth
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| | - Nicholas Waters
- Integrative Research Laboratories Sweden AB, Göteborg, Sweden (S.W., C.S., P.S., J.T., E.L., J.G., M.E., B.S., A.F., J.K., N.W.); Pharmacilitator AB, Vallda, Sweden (S.H.); Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden (S.H.); Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy (M.C.); Department of Pharmacology, Gothenburg University, Gothenburg, Sweden (S.W.); and Department of Clin Neuroscience, Karolinska Institute, Stockholm, Sweden (J.T.)
| |
Collapse
|
12
|
Bezard E. Models of hyperkinetic disorders in primates. J Neurosci Methods 2020; 332:108551. [DOI: 10.1016/j.jneumeth.2019.108551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/19/2022]
|
13
|
Zeiss CJ, Shin D, Vander Wyk B, Beck AP, Zatz N, Sneiderman CA, Kilicoglu H. Menagerie: A text-mining tool to support animal-human translation in neurodegeneration research. PLoS One 2019; 14:e0226176. [PMID: 31846471 PMCID: PMC6917268 DOI: 10.1371/journal.pone.0226176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Discovery studies in animals constitute a cornerstone of biomedical research, but suffer from lack of generalizability to human populations. We propose that large-scale interrogation of these data could reveal patterns of animal use that could narrow the translational divide. We describe a text-mining approach that extracts translationally useful data from PubMed abstracts. These comprise six modules: species, model, genes, interventions/disease modifiers, overall outcome and functional outcome measures. Existing National Library of Medicine natural language processing tools (SemRep, GNormPlus and the Chemical annotator) underpin the program and are further augmented by various rules, term lists, and machine learning models. Evaluation of the program using a 98-abstract test set achieved F1 scores ranging from 0.75-0.95 across all modules, and exceeded F1 scores obtained from comparable baseline programs. Next, the program was applied to a larger 14,481 abstract data set (2008-2017). Expected and previously identified patterns of species and model use for the field were obtained. As previously noted, the majority of studies reported promising outcomes. Longitudinal patterns of intervention type or gene mentions were demonstrated, and patterns of animal model use characteristic of the Parkinson's disease field were confirmed. The primary function of the program is to overcome low external validity of animal model systems by aggregating evidence across a diversity of models that capture different aspects of a multifaceted cellular process. Some aspects of the tool are generalizable, whereas others are field-specific. In the initial version presented here, we demonstrate proof of concept within a single disease area, Parkinson's disease. However, the program can be expanded in modular fashion to support a wider range of neurodegenerative diseases.
Collapse
Affiliation(s)
- Caroline J. Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| | - Dongwook Shin
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland, United States of America
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Amanda P. Beck
- Department of Pathology, Albert Einstein College of Medicine, New York, United States of America
| | - Natalie Zatz
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, United States of America
| | - Charles A. Sneiderman
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland, United States of America
| | - Halil Kilicoglu
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland, United States of America
| |
Collapse
|
14
|
Peng Q, Zhong S, Tan Y, Zeng W, Wang J, Cheng C, Yang X, Wu Y, Cao X, Xu Y. The Rodent Models of Dyskinesia and Their Behavioral Assessment. Front Neurol 2019; 10:1016. [PMID: 31681132 PMCID: PMC6798181 DOI: 10.3389/fneur.2019.01016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/09/2019] [Indexed: 12/24/2022] Open
Abstract
Dyskinesia, a major motor complication resulting from dopamine replacement treatment, manifests as involuntary hyperkinetic or dystonic movements. This condition poses a challenge to the treatment of Parkinson's disease. So far, several behavioral models based on rodent with dyskinesia have been established. These models have provided an important platform for evaluating the curative effect of drugs at the preclinical research level over the past two decades. However, there are differences in the modeling and behavioral testing procedures among various laboratories that adversely affect the rat and mouse models as credible experimental tools in this field. This article systematically reviews the history, the pros and cons, and the controversies surrounding rodent models of dyskinesia as well as their behavioral assessment protocols. A summary of factors that influence the behavioral assessment in the rodent dyskinesia models is also presented, including the degree of dopamine denervation, stereotaxic lesion sites, drug regimen, monitoring styles, priming effect, and individual and strain differences. Besides, recent breakthroughs like the genetic mouse models and the bilateral intoxication models for dyskinesia are also discussed.
Collapse
Affiliation(s)
- Qiwei Peng
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoping Zhong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Tan
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - WeiQi Zeng
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chi Cheng
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Fox SH, Brotchie JM. Viewpoint: Developing drugs for levodopa-induced dyskinesia in PD: Lessons learnt, what does the future hold? Eur J Neurosci 2018; 49:399-409. [PMID: 30269407 DOI: 10.1111/ejn.14173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 12/21/2022]
Abstract
The drive to develop drugs to treat PD starts and ends with the patient. Herein, we discuss how the experience with drug development for LID has led the field in translational studies in PD with advancing ground-breaking science via rigorous clinical trial design, to deliver clinical proof-of-concepts across multiple therapeutic targets. However, issues remain in advancing drugs efficacious preclinically to the clinic, and future studies need to learn from past successes and failures. Such lessons include implementing better early indicators of tolerability, for instance evaluating non-motor symptoms in preclinical models; improving patient-related outcome measures in clinical trials, as well as considering the unique nature of dyskinesia in an individual patient. The field of translational studies needs to become more patient focused to improve successful outcomes.
Collapse
Affiliation(s)
- Susan H Fox
- The Edmond J Safra Program in Parkinson Disease and Movement Disorder Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jonathan M Brotchie
- Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada.,Atuka Inc, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Modeling Parkinson’s disease and treatment complications in rodents: Potentials and pitfalls of the current options. Behav Brain Res 2018; 352:142-150. [DOI: 10.1016/j.bbr.2017.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 01/05/2023]
|
17
|
Loiodice S, Denibaud AS, Deffains W, Alix M, Montagne P, Seffals M, Drieu La Rochelle C. Validation of a New Scoring Scale for Behavioral Assessment of L-Dopa-Induced Dyskinesia in the Rat: A New Tool for Early Decision-Making in Drug Development. ACS Chem Neurosci 2018; 9:762-772. [PMID: 29226687 DOI: 10.1021/acschemneuro.7b00426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated nonhuman primate (NHP) has been described as the most translatable model for experimental reproduction of L-dopa-induced dyskinesia (LID). However, from a drug discovery perspective, the risk associated with investment in this type of model is high due to the time and cost. The 6-hydroxydopamine (6-OHDA) rat dyskinesia model is recommended for testing compounds but relies on onerous, and nonstandard behavioral rating scales. We sought to develop a simplified and sensitive method aiming at assessing LID in the rat. The purpose was to validate a reliable tool providing earlier insight into the antidyskinetic potential of compounds in a time/cost-effective manner before further investigation in NHP models. Unilaterally 6-OHDA-lesioned rats were administered L-dopa (20 mg/kg) and benserazide (5 mg/kg) daily for 3 weeks starting 4 weeks postlesion, then coadministered with amantadine (20-30-40 mg/kg). An adapted rating scale was used to score LID frequency and a severity coefficient was applied depending on the features of the observed behavior. A gradual increase (about 3-fold) in LID score was observed over the 3 weeks of L-dopa treatment. The rating scale was sensitive enough to highlight a dose-dependent amantadine-mediated decrease (about 2.2-fold) in LID score. We validated a simplified method, able to reflect different levels of severity in the assessment of LID and, thus, provide a reliable tool for drug discovery.
Collapse
Affiliation(s)
- Simon Loiodice
- Non-Clinical Department, Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Anne-Sophie Denibaud
- Non-Clinical Department, Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Wendy Deffains
- Non-Clinical Department, Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Magali Alix
- Non-Clinical Department, Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Pierre Montagne
- Non-Clinical Department, Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Marine Seffals
- Plate-Forme H2P2, Université de Rennes 1, Biosit, 2 Av. du Prof. Léon Bernard, 35043 Rennes, France
| | | |
Collapse
|
18
|
Cenci MA, Crossman AR. Animal models of l-dopa-induced dyskinesia in Parkinson's disease. Mov Disord 2018; 33:889-899. [PMID: 29488257 DOI: 10.1002/mds.27337] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/25/2022] Open
Abstract
Understanding the biological mechanisms of l-dopa-induced motor complications is dependent on our ability to investigate these phenomena in animal models of Parkinson's disease. The most common motor complications consist in wearing-off fluctuations and abnormal involuntary movements appearing when plasma levels of l-dopa are high, commonly referred to as peak-dose l-dopa-induced dyskinesia. Parkinsonian models exhibiting these features have been well-characterized in both rodent and nonhuman primate species. The first animal models of peak-dose l-dopa-induced dyskinesia were produced in monkeys lesioned with N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and treated chronically with l-dopa to elicit choreic movements and dystonic postures. Seminal studies were performed in these models using both metabolic mapping and electrophysiological techniques, providing fundamental pathophysiological insights that have stood the test of time. A decade later, it was shown possible to reproduce peak-dose l-dopa-induced dyskinesia in rats and mice rendered parkinsonian with nigrostriatal 6-hydroxydopamine lesions. When treated with l-dopa, these animals exhibit abnormal involuntary movements having both hyperkinetic and dystonic components. These models have enabled molecular- and cellular-level investigations into the mechanisms of l-dopa-induced dyskinesia. A flourishing literature using genetically engineered mice is now unraveling the role of specific genes and neural circuits in the development of l-dopa-induced motor complications. Both non-human primate and rodent models of peak-dose l-dopa-induced dyskinesia have excellent construct validity and provide valuable tools for discovering therapeutic targets and evaluating potential treatments. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- M Angela Cenci
- Department of Experimental Medical Science, Basal Ganglia Pathophysiology Unit, Lund University, Lund, Sweden
| | | |
Collapse
|
19
|
Differential Synaptic Remodeling by Dopamine in Direct and Indirect Striatal Projection Neurons in Pitx3 -/- Mice, a Genetic Model of Parkinson's Disease. J Neurosci 2018; 38:3619-3630. [PMID: 29483281 DOI: 10.1523/jneurosci.3184-17.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/23/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
In toxin-based models of Parkinson's disease (PD), striatal projection neurons (SPNs) exhibit dendritic atrophy and spine loss concurrent with an increase in excitability. Chronic l-DOPA treatment that induces dyskinesia selectively restores spine density and excitability in indirect pathway SPNs (iSPNs), whereas spine loss and hyperexcitability persist in direct pathway SPNs (dSPNs). These alterations have only been characterized in toxin-based models of PD, raising the possibility that they are an artifact of exposure to the toxin, which may engage compensatory mechanisms independent of the PD-like pathology or due to the loss of dopaminergic afferents. To test all these, we studied the synaptic remodeling in Pitx3-/- or aphakia mice, a genetic model of PD, in which most of the dopamine neurons in the substantia nigra fail to fully differentiate and to innervate the striatum. We made 3D reconstructions of the dendritic arbor and measured excitability in identified SPNs located in dorsal striatum of BAC-Pitx3-/- mice treated with saline or l-DOPA. Both dSPNs and iSPNs from BAC-Pitx3-/- mice had shorter dendritic trees, lower spine density, and more action potentials than their counterparts from WT mice. Chronic l-DOPA treatment restored spine density and firing rate in iSPNs. By contrast, in dSPNs, spine loss and hyperexcitability persisted following l-DOPA treatment, which is similar to what happens in 6-OHDA WT mice. This indicates that dopamine-mediated synaptic remodeling and plasticity is independent of dopamine innervation during SPN development and that Pitx3-/- mice are a good model because they develop the same pathology described in the toxins-based models and in human postmortem studies of advanced PD.SIGNIFICANCE STATEMENT As the only genetic model of Parkinson's disease (PD) that develops dyskinesia, Pitx3-/- mice reproduce the behavioral effects seen in humans and are a good system for studying dopamine-induced synaptic remodeling. The studies we present here establish that the structural and functional synaptic plasticity that occur in striatal projection neurons in PD and in l-DOPA-induced dyskinesia are specifically due to modulation of the neurotransmitter dopamine and are not artifacts of the use of chemical toxins in PD models. In addition, our findings provide evidence that synaptic plasticity in the Pitx3-/- mouse is similar to that seen in toxin models despite its lack of dopaminergic innervation of the striatum during development. Pitx3-/- mice reproduced the alterations described in patients with advanced PD and in well accepted toxin-based models of PD and dyskinesia. These results further consolidate the fidelity of the Pitx3-/- mouse as a PD model in which to study the morphological and physiological remodeling of striatal projection neurons by administration of l-DOPA and other drugs.
Collapse
|
20
|
Creed RB, Goldberg MS. New Developments in Genetic rat models of Parkinson's Disease. Mov Disord 2018; 33:717-729. [PMID: 29418019 DOI: 10.1002/mds.27296] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/04/2017] [Accepted: 12/10/2017] [Indexed: 12/12/2022] Open
Abstract
Preclinical research on Parkinson's disease has relied heavily on mouse and rat animal models. Initially, PD animal models were generated primarily by chemical neurotoxins that induce acute loss of dopaminergic neurons in the substantia nigra. On the discovery of genetic mutations causally linked to PD, mice were used more than rats to generate laboratory animals bearing PD-linked mutations because mutagenesis was more difficult in rats. Recent advances in technology for mammalian genome engineering and optimization of viral expression vectors have increased the use of genetic rat models of PD. Emerging research tools include "knockout" rats with disruption of genes in which mutations have been causally linked to PD, including LRRK2, α-synuclein, Parkin, PINK1, and DJ-1. Rats have also been increasingly used for transgenic and viral-mediated overexpression of genes relevant to PD, particularly α-synuclein. It may not be realistic to obtain a single animal model that completely reproduces every feature of a human disease as complex as PD. Nevertheless, compared with mice with the same mutations, many genetic rat animal models of PD better reproduce key aspects of PD including progressive loss of dopaminergic neurons in the substantia nigra, locomotor behavior deficits, and age-dependent formation of abnormal α-synuclein protein aggregates. Here we briefly review new developments in genetic rat models of PD that may have greater potential for identifying underlying mechanisms, for discovering novel therapeutic targets, and for developing greatly needed treatments to slow or halt disease progression. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rose B Creed
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Matthew S Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
21
|
Wang WW, Zhang MM, Zhang XR, Zhang ZR, Chen J, Feng L, Xie CL. A Meta-Analysis of Adenosine A2A Receptor Antagonists on Levodopa-Induced Dyskinesia In Vivo. Front Neurol 2017; 8:702. [PMID: 29375464 PMCID: PMC5770694 DOI: 10.3389/fneur.2017.00702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/06/2017] [Indexed: 12/31/2022] Open
Abstract
Background Long-term use of levodopa (l-dopa) is inevitably complicated with highly disabling fluctuations and drug-induced dyskinesias, which pose major challenges to the existing drug therapy of Parkinson's disease. Methods In this study, we conducted a systematic review and meta-analysis to assess the efficacy of A2A receptor antagonists on reducing l-dopa-induced dyskinesias (LID). Results Nine studies with a total of 152 animals were included in this meta-analysis. Total abnormal involuntary movements (AIM) score, locomotor activity, and motor disability were reported as outcome measures in 5, 5, and 3 studies, respectively. Combined standardized mean difference (SMD) estimates were calculated using a random-effects model. We pooled the whole data and found that, when compared to l-dopa alone, A2A receptor antagonists plus l-dopa treatment showed no effect on locomotor activity (SMD -0.00, 95% confidence interval (CI): -2.52 to 2.52, p = 1.0), superiority in improvement of motor disability (SMD -5.06, 95% CI: -9.25 to -0.87, p = 0.02) and more effective in control of AIM (SMD -1.82, 95% CI: -3.38 to -0.25, p = 0.02). Conclusion To sum up, these results demonstrated that A2A receptor antagonists appear to have efficacy in animal models of LID. However, large randomized clinical trials testing the effects of A2A receptor antagonists in LID patients are always warranted.
Collapse
Affiliation(s)
- Wen-Wen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Man-Man Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xing-Ru Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeng-Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Feng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng-Long Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Tronci E, Francardo V. Animal models of L-DOPA-induced dyskinesia: the 6-OHDA-lesioned rat and mouse. J Neural Transm (Vienna) 2017; 125:1137-1144. [PMID: 29242978 DOI: 10.1007/s00702-017-1825-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Abstract
Appearance of L-DOPA-induced dyskinesia (LID) represents a major limitation in the pharmacological therapy with the dopamine precursor L-DOPA. Indeed, the vast majority of parkinsonian patients develop dyskinesia within 9-10 years of L-DOPA oral administration. This makes the discovery of new therapeutic strategies an important need. In the last decades, several animal models of Parkinson's disease (PD) have been developed, to both study mechanisms underlying PD pathology and treatment-induced side effects (i.e., LID) and to screen for new potential anti-parkinsonian and anti-dyskinetic treatments. Among all the models developed, the 6-OHDA-lesioned rodents represent the models of choice to mimic PD motor symptoms and LID, thanks to their reproducibility and translational value. Under L-DOPA treatment, rodents sustaining 6-OHDA lesions develop abnormal involuntary movements with dystonic and hyperkinetic features, resembling what seen in dyskinetic PD patients. These models have been extensively validated by the evidence that dyskinetic behaviors are alleviated by compounds reducing dyskinesia in patients and non-human primate models of PD. This article will focus on the translational value of the 6-OHDA rodent models of LID, highlighting their main features, advantages and disadvantages in preclinical research.
Collapse
Affiliation(s)
- Elisabetta Tronci
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Cittadella Universitaria, SS554 Km 4.5, 09042, Monserrato, Italy.
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Goto S. Striatal Gα olf/cAMP Signal-Dependent Mechanism to Generate Levodopa-Induced Dyskinesia in Parkinson's Disease. Front Cell Neurosci 2017; 11:364. [PMID: 29201000 PMCID: PMC5696598 DOI: 10.3389/fncel.2017.00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/06/2017] [Indexed: 11/24/2022] Open
Abstract
The motor symptoms of Parkinson’s disease (PD) result from striatal dopamine (DA) deficiency due to a progressive degeneration of nigral dopaminergic cells. Although DA replacement therapy is the mainstay to treat parkinsonian symptoms, a long-term daily administration of levodopa often develops levodopa-induced dyskinesia (LID). LID is closely linked to the dysregulation of cyclic adenosine monophosphate (cAMP) signaling cascades in the medium spiny neurons (MSNs), the principal neurons of the striatum, which are roughly halved with striatonigral MSNs by striatopallidal MSNs. The olfactory type G-protein α subunit (Gαolf) represents an important regulator of the cAMP signal activities in the striatum, where it positively couples with D1-type dopamine receptor (D1R) and adenosine A2A receptor (A2AR) to increase cAMP production in the MSNs. Notably, D1Rs are primarily expressed in striatonigral MSNs, whereas D2Rs and A2ARs are expressed in striatopallidal MSNs. Based on the evidence obtained from parkinsonian mice, we hypothesized that in the DA-denervated striatum with D1R hypersensitivity, a repeated and pulsatile exposure to levodopa might cause a usage-induced degradation of Gαolf proteins in striatal MSNs, resulting in increased and decreased levels of Gαolf protein in the striatonigral and striatopallidal MSNs, respectively. As a principal cause for generating LID, this might lead to an increased responsiveness to levodopa exposure in both striatonigral and striatopallidal MSNs. Our hypothesis reinforces the long-standing concept that LID might result from the reduced activity of the striatopallidal pathway and has important clinical implications.
Collapse
Affiliation(s)
- Satoshi Goto
- Department of Neurodegenerative Disorders Research, Institute of Biomedical Sciences, Graduate School of Medical Sciences, Tokushima University, Tokushima, Japan.,Parkinson's Disease and Dystonia Research Center, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
24
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|
25
|
Bez F, Francardo V, Cenci MA. Dramatic differences in susceptibility to l-DOPA-induced dyskinesia between mice that are aged before or after a nigrostriatal dopamine lesion. Neurobiol Dis 2016; 94:213-25. [DOI: 10.1016/j.nbd.2016.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 12/26/2022] Open
|
26
|
Sebastianutto I, Maslava N, Hopkins CR, Cenci MA. Validation of an improved scale for rating l-DOPA-induced dyskinesia in the mouse and effects of specific dopamine receptor antagonists. Neurobiol Dis 2016; 96:156-170. [PMID: 27597526 DOI: 10.1016/j.nbd.2016.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 11/17/2022] Open
Abstract
Rodent models of l-DOPA-induced dyskinesia (LID) are essential to investigate pathophysiological mechanisms and treatment options. Ratings of abnormal involuntary movements (AIMs) are used to capture both qualitative and quantitative features of dyskinetic behaviors. Thus far, validated rating scales for the mouse have anchored the definition of severity to the time during which AIMs are present. Here we have asked whether the severity of axial, limb, and orolingual AIMs can be objectively assessed with scores based on movement amplitude. Mice sustained 6-OHDA lesions in the medial forebrain bundle and were treated with l-DOPA (3-6mg/kg/day) until they developed stable AIMs scores. Two independent investigators rated AIM severity using both the validated time-based scale and a novel amplitude scale, evaluating the degree of deviation of dyskinetic body parts relative to their resting position. The amplitude scale yielded a high degree of consistency both within- and between raters. Thus, time-based scores, amplitude scores, and a combination of the two ('global AIM scores') were applied to compare antidyskinetic effects produced by amantadine and by the following subtype-specific DA receptor antagonists: SCH23390 (D1/D5), Raclopride (D2/D3), PG01037 (D3), L-745,870 (D4), and VU6004461 (D4). SCH23390 and Raclopride produced similarly robust reductions in both time-based scores and amplitude scores, while PG01037 and L-745,870 had more partial effects. Interestingly, a novel and highly brain penetrable D4 receptor antagonist (VU6004461) markedly attenuated both time-based and amplitude scores without diminishing the general motor stimulant effect of l-DOPA. In summary, our results show that a dyskinesia scale combining a time dimension with an amplitude dimension ('global AIMs') is more sensitive than unidimensional scales. Moreover, the antidyskinetic effects produced by two chemically distinct D4 antagonists identify the D4 receptor as a potential future target for the treatment of LID.
Collapse
Affiliation(s)
- Irene Sebastianutto
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden.
| | - Natallia Maslava
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden.
| |
Collapse
|
27
|
Characterization of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, in rat partial and full nigral 6-hydroxydopamine lesion models of Parkinson's disease. Brain Res 2016; 1646:354-365. [DOI: 10.1016/j.brainres.2016.05.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/20/2022]
|
28
|
Debowski K, Drummer C, Lentes J, Cors M, Dressel R, Lingner T, Salinas-Riester G, Fuchs S, Sasaki E, Behr R. The transcriptomes of novel marmoset monkey embryonic stem cell lines reflect distinct genomic features. Sci Rep 2016; 6:29122. [PMID: 27385131 PMCID: PMC4935898 DOI: 10.1038/srep29122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 06/13/2016] [Indexed: 12/21/2022] Open
Abstract
Embryonic stem cells (ESCs) are useful for the study of embryonic development. However, since research on naturally conceived human embryos is limited, non-human primate (NHP) embryos and NHP ESCs represent an excellent alternative to the corresponding human entities. Though, ESC lines derived from naturally conceived NHP embryos are still very rare. Here, we report the generation and characterization of four novel ESC lines derived from natural preimplantation embryos of the common marmoset monkey (Callithrix jacchus). For the first time we document derivation of NHP ESCs derived from morula stages. We show that quantitative chromosome-wise transcriptome analyses precisely reflect trisomies present in both morula-derived ESC lines. We also demonstrate that the female ESC lines exhibit different states of X-inactivation which is impressively reflected by the abundance of the lncRNA X inactive-specific transcript (XIST). The novel marmoset ESC lines will promote basic primate embryo and ESC studies as well as preclinical testing of ESC-based regenerative approaches in NHP.
Collapse
Affiliation(s)
- Katharina Debowski
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Charis Drummer
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jana Lentes
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Maren Cors
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen (UMG), Humboldtallee 34, 37073 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Thomas Lingner
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Gabriela Salinas-Riester
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Sigrid Fuchs
- Department of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Erika Sasaki
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, 3-25-12 Tonomachi Kawasaki-ku, Kawasaki, 210-0821 Japan.,Keio Advanced Research Center, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany
| |
Collapse
|
29
|
Huang L, Merson TD, Bourne JA. In vivo whole brain, cellular and molecular imaging in nonhuman primate models of neuropathology. Neurosci Biobehav Rev 2016; 66:104-18. [PMID: 27151822 DOI: 10.1016/j.neubiorev.2016.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/31/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Rodents have been the principal model to study brain anatomy and function due to their well-mapped brain architecture, rapid reproduction and amenability to genetic modification. However, there are clear limitations, for example their simpler neocortex, necessitating the need to adopt a model that is closer to humans in order to understand human cognition and brain conditions. Nonhuman primates (NHPs) are ideally suited as they are our closest relatives in the animal kingdom but in vivo imaging technologies to study brain structure and function in these species can be challenging. With the surge in NHP research in recent years, scientists have begun adapting imaging technologies, such as two-photon microscopy, for these species. Here we review the various NHP models that exist as well as their use in advanced microscopic and mesoscopic studies. We discuss the challenges in the field and investigate the opportunities that lie ahead.
Collapse
Affiliation(s)
- Lieven Huang
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia
| | - Tobias D Merson
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria, 3052, Australia
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia.
| |
Collapse
|
30
|
Gupta DK, Hang X, Liu R, Hasan A, Feng Z. Levodopa-Induced Motor and Dopamine Receptor Changes in Caenorhabditis elegans Overexpressing Human Alpha-Synuclein. NEURODEGENER DIS 2015; 16:179-83. [PMID: 26606044 DOI: 10.1159/000440845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/03/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Levodopa-induced dyskinesia (LID) is a disabling complication of levodopa therapy in Parkinson's disease (PD) with no effective treatments. Fluctuations in levels of levodopa constitute a key risk factor of LID. There is a pressing need for the development of a simple animal model of LID. Several genetic and toxin-based models of PD in Caenorhabditis elegans have been described, which have advanced our understanding of PD pathophysiology. We aimed to study levodopa-induced changes in a Parkinson's disease model of C. elegans expressing human α-synuclein. METHODS We exposed the α-synuclein C. elegans to levodopa in continuous and alternating fashions. Automated behavioral analysis was then used to quantify changes in motor activity. Confocal microscopy was used next to quantify changes in dopamine receptor distribution and expression in motor neurons of live C. elegans. RESULTS Chronic exposure to levodopa led to hyperactivity of the α-synuclein C. elegans without meaningful increase in motor activity. There was also an increase in peripheral clustering and expression of dopamine receptors in motor neurons. Both of these changes were significantly higher with alternating, compared to continuous, exposure to levodopa. CONCLUSIONS This is the first report of changes in motor and dopamine receptors induced by levodopa in C. elegans overexpressing human α-synuclein. We propose that these phenotypes represent a simple animal model of LID in C. elegans. Such a model holds the promise of enabling high-throughput screenings for potential therapeutic targets and drug candidates.
Collapse
Affiliation(s)
- Deepak K Gupta
- Department of Neurology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
31
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 348] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
32
|
Fieblinger T, Cenci MA. Zooming in on the small: the plasticity of striatal dendritic spines in L-DOPA-induced dyskinesia. Mov Disord 2015; 30:484-93. [PMID: 25759263 DOI: 10.1002/mds.26139] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/07/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
The spiny dendrites of striatal projection neurons integrate synaptic inputs of different origins to regulate movement. It has long been known that these dendrites lose spines and display atrophic features in Parkinson's disease (PD), but the significance of these morphological changes has remained unknown. Some recent studies reveal a remarkable structural plasticity of striatal spines in parkinsonian rodents treated with L-3,4-dihydroxyphenylalanine (L-DOPA), and they demonstrate an association between this plasticity and the development of dyskinesia. These studies used different approaches and animal models, which possibly explains why they emphasize different plastic changes as being most closely linked to dyskinesia (such as a growth of new spines in neurons of the indirect pathway, or a loss of spines in neurons of the direct pathway, or the appearance of spines with aberrant synaptic features). Clearly, further investigations are required to reconcile these intriguing findings and integrate them in a coherent pathophysiological model. Nevertheless, these studies may mark the beginning of a new era for dyskinesia research. In addition to addressing neurochemical and molecular events that trigger involuntary movements, there is a need to better understand the long-lasting structural reorganization of cells and circuits that maintain the brain in a "dyskinesia-prone" state. This may lead to the identification of new efficacious approaches to prevent the complications of dopaminergic therapies in PD.
Collapse
Affiliation(s)
- Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC F11, 221 84 Lund, Sweden
| | | |
Collapse
|
33
|
Iderberg H, Maslava N, Thompson AD, Bubser M, Niswender CM, Hopkins CR, Lindsley CW, Conn PJ, Jones CK, Cenci MA. Pharmacological stimulation of metabotropic glutamate receptor type 4 in a rat model of Parkinson's disease and L-DOPA-induced dyskinesia: Comparison between a positive allosteric modulator and an orthosteric agonist. Neuropharmacology 2015; 95:121-9. [PMID: 25749357 DOI: 10.1016/j.neuropharm.2015.02.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/16/2015] [Accepted: 02/18/2015] [Indexed: 12/21/2022]
Abstract
Metabotropic glutamate receptor 4 (mGlu4) negatively modulates GABA and glutamate release in the 'indirect pathway' of the basal ganglia, and has thus been proposed as a potential target to treat motor symptoms in Parkinson's disease. Here, we present an extensive comparison of the behavioural effects produced by the mGlu4 positive allosteric modulator (PAM), VU0364770, and the mGlu4 orthosteric agonist, LSP1-2111, in rats with unilateral 6-OHDA lesions. The compounds' activity was initially assessed in a test of haloperidol-induced catalepsy in intact rats, and effective doses were then evaluated in the hemiparkinsonian animal model. Neither of the two compounds modified the development of dyskinetic behaviours elicited by chronic treatment with full doses of l-DOPA. When given together with l-DOPA to rats with already established dyskinesias, neither VU0364770 nor LSP1-2111 modified the abnormal involuntary movement scores. VU0364770 potentiated, however, the motor stimulant effect of a subthreshold l-DOPA dose in certain behavioural tests, whereas LSP1-2111 lacked this ability. Taken together, these results indicate that a pharmacological stimulation of mGlu4 lacks intrinsic antidyskinetic activity, but may have DOPA-sparing activity in Parkinson's disease. For the latter indication, mGlu4 PAMs appear to provide a better option than orthosteric agonists.
Collapse
Affiliation(s)
- Hanna Iderberg
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Sweden.
| | - Natallia Maslava
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Sweden
| | - Analisa D Thompson
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Corey R Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Sweden.
| |
Collapse
|
34
|
Potts LF, Uthayathas S, Greven ACM, Dyavarshetty B, Mouradian MM, Papa SM. A new quantitative rating scale for dyskinesia in nonhuman primates. Behav Pharmacol 2015; 26:109-16. [PMID: 25171151 PMCID: PMC4276436 DOI: 10.1097/fbp.0000000000000084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The aim of this study was to develop a quantitative scale to assess levodopa-induced dyskinesias (LIDs) in nonhuman primates using a video-based scoring system [Quantitative Dyskinesia Scale (QDS)]. Six macaques with stable Parkinsonism and LID were used for tests of the new QDS, in comparison with our current standardized scale (Drug-Related Side effects), which provides a classic subjective measurement of dyskinesia. QDS scoring is based on systematic movement counts in time frames, using videotape recordings. For both scales, body segments scored included each extremity, the trunk, the neck, and the face, and raters were blinded to L-dopa treatments. Comparison of the two scales revealed that their scores are highly correlated with and are parallel to the L-dopa pharmacokinetic profile, although the QDS provided significantly more quantifiable measurements. This remained the case after separating animals into groups of mild and severe dyskinesias. Inter-rater reliability for application of the QDS was confirmed from scores obtained by three examiners. We conclude that the QDS is a quantitative tool for reliably scoring LID in parkinsonian monkeys at all levels of severity of dyskinesia. The application of this new standard for scoring LID in primates will allow for more precise measurements of the effects of experimental treatments and will improve the quality of results obtained in translational studies.
Collapse
Affiliation(s)
- Lisa F Potts
- aDivision of Neuropharmacology and Neurologic Diseases bDepartment of Neurology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia cDepartment of Neurology, Center for Neurodegenerative and Neuroimmunologic Diseases, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | | | | | | | | | | |
Collapse
|
35
|
Rodent Models of Treatment-Related Complications in Parkinson Disease. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00022-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
36
|
Quik M, Bordia T, Zhang D, Perez XA. Nicotine and Nicotinic Receptor Drugs. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 124:247-71. [DOI: 10.1016/bs.irn.2015.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
37
|
Cenci MA. Presynaptic Mechanisms of l-DOPA-Induced Dyskinesia: The Findings, the Debate, and the Therapeutic Implications. Front Neurol 2014; 5:242. [PMID: 25566170 PMCID: PMC4266027 DOI: 10.3389/fneur.2014.00242] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/10/2014] [Indexed: 12/24/2022] Open
Abstract
The dopamine (DA) precursor l-DOPA has been the most effective treatment for Parkinson’s disease (PD) for over 40 years. However, the response to this treatment changes with disease progression, and most patients develop dyskinesias (abnormal involuntary movements) and motor fluctuations within a few years of l-DOPA therapy. There is wide consensus that these motor complications depend on both pre- and post-synaptic disturbances of nigrostriatal DA transmission. Several presynaptic mechanisms converge to generate large DA swings in the brain concomitant with the peaks-and-troughs of plasma l-DOPA levels, while post-synaptic changes engender abnormal functional responses in dopaminoceptive neurons. While this general picture is well-accepted, the relative contribution of different factors remains a matter of debate. A particularly animated debate has been growing around putative players on the presynaptic side of the cascade. To what extent do presynaptic disturbances in DA transmission depend on deficiency/dysfunction of the DA transporter, aberrant release of DA from serotonin neurons, or gliovascular mechanisms? And does noradrenaline (which is synthetized from DA) play a role? This review article will summarize key findings, controversies, and pending questions regarding the presynaptic mechanisms of l-DOPA-induced dyskinesia. Intriguingly, the debate around these mechanisms has spurred research into previously unexplored facets of brain plasticity that have far-reaching implications to the treatment of neuropsychiatric disease.
Collapse
Affiliation(s)
- M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University , Lund , Sweden
| |
Collapse
|
38
|
Meng T, Yuan S, Zheng Z, Liu T, Lin L. Effects of endogenous melatonin on glutamate and GABA rhythms in the striatum of unilateral 6-hydroxydopamine-lesioned rats. Neuroscience 2014; 286:308-15. [PMID: 25499317 DOI: 10.1016/j.neuroscience.2014.11.062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 10/24/2022]
Abstract
We have previously reported a time-dependent increase in melatonin (MLT) and decrease in dopamine (DA) in striatal dialysate 3 weeks after unilateral 6-hydroxydopamine (6-OHDA) lesioning in the rat substantia nigra pars compacta (SNc) and medial forebrain bundle (MFB). This study aimed to investigate dynamic and circadian variations in DA, MLT, glutamate (Glu) and γ-aminobutyric acid (GABA) in striatal dialysates in the same 6-OHDA animal model. These neurotransmitters were determined using high-performance liquid chromatography (HPLC). Three weeks following 6-OHDA lesioning, there was a significant increase in extracellular Glu (156%) and decrease in GABA (15%) and DA (85%) in the lesioned striatum. These changes continued over time. Concomitantly, MLT was increased by 107% in the lesioned striatal dialysates after 4 weeks, and continued to increase gradually over time. Six weeks post-treatment, levels of MLT secretion at 12 time points were higher, and the peak time of MLT secretion was earlier, in 6-OHDA-lesioned rats compared with vehicle-treated rats. In addition, significant variations in extracellular levels of Glu and GABA between day and night were observed in vehicle-treated rat striatum. However, no circadian variations were observed in the striatum of unilateral 6-OHDA-lesioned rats. Six weeks post-treatment, MLT levels correlated well with Glu and GABA levels at corresponding time-points in the striatum ipsilateral to the injected side in both groups, and increased MLT levels also correlated well with changes in Glu and GABA in the striatum in 6-OHDA-lesioned rats. These data suggest that 6-OHDA lesioning affects the endogenous productions of DA, MLT, Glu and GABA, and changes the MLT secretion pattern. Augmented striatal MLT levels and advanced MLT secretion pattern caused by unilateral intracerebral injection of 6-OHDA may influence the variations in Glu and GABA between day and night.
Collapse
Affiliation(s)
- T Meng
- Research Center of Neurobiology, Fujian Medical University, Fuzhou 350108, China; Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China.
| | - S Yuan
- Department of Neurosurgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| | - Z Zheng
- Research Center of Neurobiology, Fujian Medical University, Fuzhou 350108, China; Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350108, China.
| | - T Liu
- Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350108, China.
| | - L Lin
- Research Center of Neurobiology, Fujian Medical University, Fuzhou 350108, China; Department of Biochemistry and Molecular Biology, Fujian Medical University, Fuzhou 350108, China.
| |
Collapse
|
39
|
Mosharov EV, Borgkvist A, Sulzer D. Presynaptic effects of levodopa and their possible role in dyskinesia. Mov Disord 2014; 30:45-53. [PMID: 25450307 DOI: 10.1002/mds.26103] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/28/2014] [Accepted: 11/01/2014] [Indexed: 12/25/2022] Open
Abstract
Levodopa replacement therapy has long provided the most effective treatment for Parkinson's disease (PD). We review how this dopamine (DA) precursor enhances dopaminergic transmission by providing a greater sphere of neurotransmitter influence as a result of the confluence of increased quantal size and decreased DA reuptake, as well as loading DA as a false transmitter into surviving serotonin neuron synaptic vesicles. We further review literature on how presynaptic dysregulation of DA release after l-dopa might trigger dyskinesias in PD patients.
Collapse
Affiliation(s)
- Eugene V Mosharov
- Departments of Neurology, Columbia University Medical Center, New York, New York, USA
| | | | | |
Collapse
|
40
|
Solís O, Espadas I, Del-Bel EA, Moratalla R. Nitric oxide synthase inhibition decreases l-DOPA-induced dyskinesia and the expression of striatal molecular markers in Pitx3(-/-) aphakia mice. Neurobiol Dis 2014; 73:49-59. [PMID: 25281315 DOI: 10.1016/j.nbd.2014.09.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/12/2014] [Accepted: 09/21/2014] [Indexed: 01/09/2023] Open
Abstract
Nitric oxide (NO), a gaseous messenger molecule synthesized by nitric oxide synthase (NOS), plays a pivotal role in integrating dopamine transmission in the basal ganglia and has been implicated in the pathogenesis of Parkinson disease (PD). To study the role of the nitrergic system in l-DOPA-induced dyskinesia (LID), we assessed the effect of the pharmacological manipulation of NO levels and NO/cyclic guanosine monophosphate (cGMP) signaling on LID in the Pitx3(-/-) aphakia mouse, a genetic model of PD. To evaluate the effect of decreased NO signaling on the development of LID, Pitx3(-/-) mice were chronically treated with l-DOPA and 7-nitroindazole (7-NI, a neuronal NOS inhibitor). To evaluate its effect on the expression of established LID, 7-NI was administered acutely to dyskinetic mice. The chronic 7-NI treatment attenuated the development of LID in the Pitx3(-/-) mice, and the sub-acute 7-NI treatment attenuated established dyskinesia without affecting the beneficial therapeutic effect of l-DOPA. Moreover, 7-NI significantly reduced FosB and pAcH3 expression in the acutely and chronically l-DOPA-treated mice. We also examined how increasing NO/cGMP signaling affects LID expression by acutely administering molsidomine (an NO donor) or zaprinast (a cGMP phosphodiesterase 5-PDE5 inhibitor) before l-DOPA in mice with established dyskinesia. Paradoxically, the administration of either of these drugs also significantly diminished the expression of established LID; however, the effect occurred at the expense of the antiparkinsonian l-DOPA properties. We demonstrate that targeting the NO/cGMP signaling pathway reduces dyskinetic behaviors and molecular markers, but only the 7-NI treatment preserved the antiparkinsonian effect of l-DOPA, indicating that NOS inhibitors represent a potential therapy to reduce LID.
Collapse
Affiliation(s)
- Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Espadas
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Elaine A Del-Bel
- Department of Morphology, Physiology and Pathology, School of Odontology, University of Sao Paulo, Campus Ribeirao Preto, Brazil
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
41
|
Mitogen- and stress-activated protein kinase 1 is required for specific signaling responses in dopamine-denervated mouse striatum, but is not necessary for L-DOPA-induced dyskinesia. Neurosci Lett 2014; 583:76-80. [PMID: 25233866 DOI: 10.1016/j.neulet.2014.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/14/2014] [Accepted: 09/08/2014] [Indexed: 11/20/2022]
Abstract
In advanced Parkinson's disease, l-DOPA treatment causes the appearance of abnormal involuntary movements or l-DOPA-induced dyskinesia (LID). LID results in part from l-DOPA-induced activation of extracellular signal-regulated kinase (ERK) in the dopamine-denervated striatum. Activated ERK triggers nuclear responses, including phosphorylation of mitogen- and stress-activated protein kinase 1 (MSK1) and histone H3, and transcription of genes such as FosB. To determine the role of MSK1, wild type and MSK1 knockout mice with unilateral 6-hydroxydopamine lesion in the dorsolateral striatum were chronically treated with l-DOPA. The absence of MSK1 had no effect on the lesion or l-DOPA-induced ERK activation, but reduced l-DOPA-induced phosphorylation of histone H3 and FosB accumulation in the dopamine-denervated striatum. MSK1 deficiency also prevented the increase in Gαolf, the stimulatory α subunit of G protein coupling striatal dopamine D1 receptor to adenylyl cyclase. However, the intensity of LID was similar in MSK1-deficient and wild type mice. In conclusion, l-DOPA-induced activation of MSK1 contributes to histone H3 phosphorylation, induction of FosB, and Gαolf up-regulation but appears not to be necessary for the development of LID.
Collapse
|
42
|
Quik M, Zhang D, Perez XA, Bordia T. Role for the nicotinic cholinergic system in movement disorders; therapeutic implications. Pharmacol Ther 2014; 144:50-9. [PMID: 24836728 DOI: 10.1016/j.pharmthera.2014.05.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 01/04/2023]
Abstract
A large body of evidence using experimental animal models shows that the nicotinic cholinergic system is involved in the control of movement under physiological conditions. This work raised the question whether dysregulation of this system may contribute to motor dysfunction and whether drugs targeting nicotinic acetylcholine receptors (nAChRs) may be of therapeutic benefit in movement disorders. Accumulating preclinical studies now show that drugs acting at nAChRs improve drug-induced dyskinesias. The general nAChR agonist nicotine, as well as several nAChR agonists (varenicline, ABT-089 and ABT-894), reduces l-dopa-induced abnormal involuntary movements or dyskinesias up to 60% in parkinsonian nonhuman primates and rodents. These dyskinesias are potentially debilitating abnormal involuntary movements that arise as a complication of l-dopa therapy for Parkinson's disease. In addition, nicotine and varenicline decrease antipsychotic-induced abnormal involuntary movements in rodent models of tardive dyskinesia. Antipsychotic-induced dyskinesias frequently arise as a side effect of chronic drug treatment for schizophrenia, psychosis and other psychiatric disorders. Preclinical and clinical studies also show that the nAChR agonist varenicline improves balance and coordination in various ataxias. Lastly, nicotine has been reported to attenuate the dyskinetic symptoms of Tourette's disorder. Several nAChR subtypes appear to be involved in these beneficial effects of nicotine and nAChR drugs including α4β2*, α6β2* and α7 nAChRs (the asterisk indicates the possible presence of other subunits in the receptor). Overall, the above findings, coupled with nicotine's neuroprotective effects, suggest that nAChR drugs have potential for future drug development for movement disorders.
Collapse
Affiliation(s)
- Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA.
| | - Danhui Zhang
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Xiomara A Perez
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| |
Collapse
|
43
|
Lin M, Zhao D, Hrabovsky A, Pedrosa E, Zheng D, Lachman HM. Heat shock alters the expression of schizophrenia and autism candidate genes in an induced pluripotent stem cell model of the human telencephalon. PLoS One 2014; 9:e94968. [PMID: 24736721 PMCID: PMC3988108 DOI: 10.1371/journal.pone.0094968] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/21/2014] [Indexed: 01/08/2023] Open
Abstract
Schizophrenia (SZ) and autism spectrum disorders (ASD) are highly heritable neuropsychiatric disorders, although environmental factors, such as maternal immune activation (MIA), play a role as well. Cytokines mediate the effects of MIA on neurogenesis and behavior in animal models. However, MIA stimulators can also induce a febrile reaction, which could have independent effects on neurogenesis through heat shock (HS)-regulated cellular stress pathways. However, this has not been well-studied. To help understand the role of fever in MIA, we used a recently described model of human brain development in which induced pluripotent stem cells (iPSCs) differentiate into 3-dimensional neuronal aggregates that resemble a first trimester telencephalon. RNA-seq was carried out on aggregates that were heat shocked at 39°C for 24 hours, along with their control partners maintained at 37°C. 186 genes showed significant differences in expression following HS (p<0.05), including known HS-inducible genes, as expected, as well as those coding for NGFR and a number of SZ and ASD candidates, including SMARCA2, DPP10, ARNT2, AHI1 and ZNF804A. The degree to which the expression of these genes decrease or increase during HS is similar to that found in copy loss and copy gain copy number variants (CNVs), although the effects of HS are likely to be transient. The dramatic effect on the expression of some SZ and ASD genes places HS, and perhaps other cellular stressors, into a common conceptual framework with disease-causing genetic variants. The findings also suggest that some candidate genes that are assumed to have a relatively limited impact on SZ and ASD pathogenesis based on a small number of positive genetic findings, such as SMARCA2 and ARNT2, may in fact have a much more substantial role in these disorders - as targets of common environmental stressors.
Collapse
Affiliation(s)
- Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Dejian Zhao
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Anastasia Hrabovsky
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail: (HML); (D. Zheng)
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail: (HML); (D. Zheng)
| |
Collapse
|
44
|
Neuroprotective Effects of Jitai Tablet, a Traditional Chinese Medicine, on the MPTP-Induced Acute Model of Parkinson's Disease: Involvement of the Dopamine System. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:542383. [PMID: 24799940 PMCID: PMC3996930 DOI: 10.1155/2014/542383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/22/2014] [Accepted: 01/29/2014] [Indexed: 01/07/2023]
Abstract
Jitai tablet (JTT) is a traditional Chinese medicine used to treat neuropsychiatric disorders. We previously demonstrated that JTT treatment led to increased level of dopamine transporter (DAT) in the striatum, thus indicating that JTT might have therapeutic potential for Parkinson's disease (PD), which is characterized by dysregulated dopamine (DA) transmission and decreased striatal DAT expression. The aim of this study was to investigate the neuroprotective effect of JTT on MPTP-induced PD mice. Using locomotor activity test and rotarod test, we evaluated the effects of JTT (0.50, 0.15, or 0.05 g/kg) on MPTP-induced behavioral impairments. Tyrosine hydroxylase TH-positive neurons in the substantia nigra and DAT and dopamine D2 receptor (D2R) levels in the striatum were detected by immunohistochemical staining and/or autoradiography. Levels of DA and its metabolites were determined by HPLC. In MPTP-treated mice, behavioral impairments were alleviated by JTT treatment. Moreover, JTT protected against impairment of TH-positive neurons and attenuated the MPTP-induced decreases in DAT and D2R. Finally, high dose of JTT (0.50 g/kg) inhibited the MPTP-induced increase in DA metabolism rate. Taken together, results from our present study provide evidence that JTT offers neuroprotective effects against the neurotoxicity of MPTP and thus might be a potential treatment for PD.
Collapse
|
45
|
Miguelez C, Morera-Herreras T, Torrecilla M, Ruiz-Ortega JA, Ugedo L. Interaction between the 5-HT system and the basal ganglia: functional implication and therapeutic perspective in Parkinson's disease. Front Neural Circuits 2014; 8:21. [PMID: 24672433 PMCID: PMC3955837 DOI: 10.3389/fncir.2014.00021] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/27/2014] [Indexed: 01/15/2023] Open
Abstract
The neurotransmitter serotonin (5-HT) has a multifaceted function in the modulation of information processing through the activation of multiple receptor families, including G-protein-coupled receptor subtypes (5-HT1, 5-HT2, 5-HT4-7) and ligand-gated ion channels (5-HT3). The largest population of serotonergic neurons is located in the midbrain, specifically in the raphe nuclei. Although the medial and dorsal raphe nucleus (DRN) share common projecting areas, in the basal ganglia (BG) nuclei serotonergic innervations come mainly from the DRN. The BG are a highly organized network of subcortical nuclei composed of the striatum (caudate and putamen), subthalamic nucleus (STN), internal and external globus pallidus (or entopeduncular nucleus in rodents, GPi/EP and GPe) and substantia nigra (pars compacta, SNc, and pars reticulata, SNr). The BG are part of the cortico-BG-thalamic circuits, which play a role in many functions like motor control, emotion, and cognition and are critically involved in diseases such as Parkinson's disease (PD). This review provides an overview of serotonergic modulation of the BG at the functional level and a discussion of how this interaction may be relevant to treating PD and the motor complications induced by chronic treatment with L-DOPA.
Collapse
Affiliation(s)
- Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU Leioa, Spain ; Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU Vitoria-Gasteiz, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU Leioa, Spain
| | - Maria Torrecilla
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU Leioa, Spain
| | - Jose A Ruiz-Ortega
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU Leioa, Spain ; Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU Vitoria-Gasteiz, Spain
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU Leioa, Spain
| |
Collapse
|
46
|
Francardo V, Cenci MA. Investigating the molecular mechanisms of L-DOPA-induced dyskinesia in the mouse. Parkinsonism Relat Disord 2014; 20 Suppl 1:S20-2. [DOI: 10.1016/s1353-8020(13)70008-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
47
|
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. To date, there is no effective treatment that halts its progression. Increasing evidence indicates that mitochondria play an important role in the development of PD. Hence mitochondria-targeted approaches or agents may have therapeutic promise for treatment of the disease. Neuropeptide CART (cocaine-amphetamine-regulated transcript), a hypothalamus and midbrain enriched neurotransmitter with an antioxidant property, can be found in mitochondria, which is the main source of reactive oxygen species. Systemic administration of CART has been found to ameliorate dopaminergic neuronal loss and improve motor functions in a mouse model of PD. In this article, we summarize recent progress in studies investigating the relationship between CART, dopamine, and the pathophysiology of PD, with a focus on mitochondria-related topics.
Collapse
|
48
|
Zhu K, van Hilten JJ, Putter H, Marinus J. Risk factors for hallucinations in Parkinson's disease: results from a large prospective cohort study. Mov Disord 2013; 28:755-62. [PMID: 23520046 DOI: 10.1002/mds.25389] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 01/04/2013] [Accepted: 01/15/2013] [Indexed: 11/11/2022] Open
Abstract
The aim of this study was to identify risk factors for the development of hallucinations in patients with Parkinson's disease (PD). A broad range of motor and nonmotor features was assessed at baseline and during the following 5 years in 386 PD patients. Cross-sectional analyses of baseline data and longitudinal analyses of follow-up data were performed to identify risk factors for hallucinations in PD. Twenty-one percent of the patients had hallucinations at baseline, whereas 46% of the patients without hallucinations at baseline developed this feature during follow-up. Univariate survival analysis showed that older age, female sex, less education, higher age at onset, and more severe motor and cognitive impairment, depression, daytimes sleepiness, autonomic dysfunction, and motor fluctuations and dyskinesias, as well as higher daily levodopa dose, were associated with the risk of developing hallucinations. This largely corresponds with the features that were associated with the presence of hallucinations at baseline. In a stepwise regression model, older age at onset, female sex, excessive daytime sleepiness, autonomic dysfunction, and dyskinesias emerged as independent risk factors for developing hallucinations. Female sex, autonomic dysfunction, motor fluctuations, and dyskinesias have not been reported as risk factors in previous studies. These findings lend support to the notion that hallucinations in PD are caused by a combination of risk factors that are associated with (the interaction between) older age and more advanced disease. The identification of female sex as a risk factor for developing of hallucinations in PD is a new finding and should be verified in future studies.
Collapse
Affiliation(s)
- Kangdi Zhu
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
49
|
Yang X, Wu N, Song L, Liu Z. Intrastriatal injections of KN-93 ameliorates levodopa-induced dyskinesia in a rat model of Parkinson's disease. Neuropsychiatr Dis Treat 2013; 9:1213-20. [PMID: 23983471 PMCID: PMC3751461 DOI: 10.2147/ndt.s45422] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Levodopa remains the most effective drug for the treatment of Parkinson's disease (PD). However, long-term levodopa treatment is associated with the emergence of levodopa-induced dyskinesia (LID), which has hampered its use for PD treatment. The mechanisms of LID are only partially understood. A previous study showed that KN-93, a Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) inhibitor, could be used to ameliorate LID in rats. However, the precise mechanisms by which KN-93 acts as an antidyskinetic are not fully understood. METHODS In the present study, a rat model of PD was induced by 6-hydroxydopamine (OHDA) injections. Then, the successfully lesioned rats were intrastriatally administered with a different dose of KN-93 (1 μg, 2 μg, or 5 μg) prior to levodopa treatment. Abnormal involuntary movements (AIMs) scores and apomorphine-induced rotations were measured in PD rats. Phosphorylated levels of GluR1 at Serine-845 (pGluR1S845) levels were determined by western blot. Arc and Penk levels were measured by real-time polymerase chain reaction (PCR). RESULTS We found that both 2 μg and 5 μg KN-93 treatment lowered AIMs scores in levodopa priming PD rats without affecting the antiparkinsonian effect of levodopa. In agreement with behavioral analysis, KN-93 treatment (2 μg) reduced pGluR1S845 levels in PD rats. Moreover, KN-93 treatment (2 μg) reduced the expression of Gad1 and Nur77 in PD rats. CONCLUSION These data indicated that intrastriatal injections of KN-93 were beneficial in reducing the expression of LID by lowering the expression of pGluR1S845 via suppressing the activation of CaMKII in PD rats. Decreased expression of pGluR1S845 further reduced the expression of Gad1 and Nur77 in PD rats.
Collapse
Affiliation(s)
- Xinxin Yang
- Department of Neurology, Xinhua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|