1
|
Ahmed MR, Zheng C, Dunning JL, Ahmed MS, Ge C, Pair FS, Gurevich VV, Gurevich EV. Arrestin-3-assisted activation of JNK3 mediates dopaminergic behavioral sensitization. Cell Rep Med 2024; 5:101623. [PMID: 38936368 PMCID: PMC11293330 DOI: 10.1016/j.xcrm.2024.101623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/15/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
In rodents with unilateral ablation of neurons supplying dopamine to the striatum, chronic treatment with the dopamine precursor L-DOPA induces a progressive increase of behavioral responses, a process known as behavioral sensitization. This sensitization is blunted in arrestin-3 knockout mice. Using virus-mediated gene delivery to the dopamine-depleted striatum of these mice, we find that the restoration of arrestin-3 fully rescues behavioral sensitization, whereas its mutant defective in c-Jun N-terminal kinase (JNK) activation does not. A 25-residue arrestin-3-derived peptide that facilitates JNK3 activation in cells, expressed ubiquitously or selectively in direct pathway striatal neurons, also fully rescues sensitization, whereas an inactive homologous arrestin-2-derived peptide does not. Behavioral rescue is accompanied by the restoration of JNK3 activity, as reflected by JNK-dependent phosphorylation of the transcription factor c-Jun in the dopamine-depleted striatum. Thus, arrestin-3-assisted JNK3 activation in direct pathway neurons is a critical element of the molecular mechanism underlying sensitization upon dopamine depletion and chronic L-DOPA treatment.
Collapse
Affiliation(s)
- Mohamed R Ahmed
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA; University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA; The University of Alabama at Birmingham, SHEL 121, 1825 University Boulevard, Birmingham, AL 35294-2182, USA
| | - Chen Zheng
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Jeffery L Dunning
- Contet Laboratory, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Mohamed S Ahmed
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Connie Ge
- University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - F Sanders Pair
- The University of Alabama at Birmingham, SHEL 121, 1825 University Boulevard, Birmingham, AL 35294-2182, USA
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA.
| |
Collapse
|
2
|
Bernardes CP, Lopes Pinheiro E, Ferreira IG, de Oliveira IS, dos Santos NAG, Sampaio SV, Arantes EC, dos Santos AC. Fraction of C. d. collilineatus venom containing crotapotin protects PC12 cells against MPP + toxicity by activating the NGF-signaling pathway. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230056. [PMID: 38915449 PMCID: PMC11194915 DOI: 10.1590/1678-9199-jvatitd-2023-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/08/2024] [Indexed: 06/26/2024] Open
Abstract
Background Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. There is no effective treatment for neurodegenerative diseases. Snake venoms are a cocktail of proteins and peptides with great therapeutic potential and might be useful in the treatment of neurodegenerative diseases. Crotapotin is the acid chain of crotoxin, the major component of Crotalus durissus collilineatus venom. PD is characterized by low levels of neurotrophins, and synaptic and axonal degeneration; therefore, neurotrophic compounds might delay the progression of PD. The neurotrophic potential of crotapotin has not been studied yet. Methods We evaluated the neurotrophic potential of crotapotin in untreated PC12 cells, by assessing the induction of neurite outgrowth. The activation of the NGF signaling pathway was investigated through pharmacological inhibition of its main modulators. Additionally, its neuroprotective and neurorestorative effects were evaluated by assessing neurite outgrowth and cell viability in PC12 cells treated with the dopaminergic neurotoxin MPP+ (1-methyl-4-phenylpyridinium), known to induce Parkinsonism in humans and animal models. Results Crotapotin induced neuritogenesis in PC12 cells through the NGF-signaling pathway, more specifically, by activating the NGF-selective receptor trkA, and the PI3K/Akt and the MAPK/ERK cascades, which are involved in neuronal survival and differentiation. In addition, crotapotin had no cytotoxic effect and protected PC12 cells against the inhibitory effects of MPP+ on cell viability and differentiation. Conclusion These findings show, for the first time, that crotapotin has neurotrophic/neuroprotective/neurorestorative potential and might be beneficial in Parkinson's disease. Additional studies are necessary to evaluate the toxicity of crotapotin in other cell models.
Collapse
Affiliation(s)
- Carolina Petri Bernardes
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Ernesto Lopes Pinheiro
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isabela Gobbo Ferreira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isadora Sousa de Oliveira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| |
Collapse
|
3
|
Evers J, Orłowski J, Jahns H, Lowery MM. On-Off and Proportional Closed-Loop Adaptive Deep Brain Stimulation Reduces Motor Symptoms in Freely Moving Hemiparkinsonian Rats. Neuromodulation 2024; 27:476-488. [PMID: 37245140 DOI: 10.1016/j.neurom.2023.03.018] [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: 12/19/2022] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVES Closed-loop adaptive deep brain stimulation (aDBS) continuously adjusts stimulation parameters, with the potential to improve efficacy and reduce side effects of deep brain stimulation (DBS) for Parkinson's disease (PD). Rodent models can provide an effective platform for testing aDBS algorithms and establishing efficacy before clinical investigation. In this study, we compare two aDBS algorithms, on-off and proportional modulation of DBS amplitude, with conventional DBS in hemiparkinsonian rats. MATERIALS AND METHODS DBS of the subthalamic nucleus (STN) was delivered wirelessly in freely moving male and female hemiparkinsonian (N = 7) and sham (N = 3) Wistar rats. On-off and proportional aDBS, based on STN local field potential beta power, were compared with conventional DBS and three control stimulation algorithms. Behavior was assessed during cylinder tests (CT) and stepping tests (ST). Successful model creation was confirmed via apomorphine-induced rotation test and Tyrosine Hydroxylase-immunocytochemistry. Electrode location was histologically confirmed. Data were analyzed using linear mixed models. RESULTS Contralateral paw use in parkinsonian rats was reduced to 20% and 25% in CT and ST, respectively. Conventional, on-off, and proportional aDBS significantly improved motor function, restoring contralateral paw use to approximately 45% in both tests. No improvement in motor behavior was observed with either randomly applied on-off or low-amplitude continuous stimulation. Relative STN beta power was suppressed during DBS. Relative power in the alpha and gamma bands decreased and increased, respectively. Therapeutically effective adaptive DBS used approximately 40% less energy than did conventional DBS. CONCLUSIONS Adaptive DBS, using both on-off and proportional control schemes, is as effective as conventional DBS in reducing motor symptoms of PD in parkinsonian rats. Both aDBS algorithms yield substantial reductions in stimulation power. These findings support using hemiparkinsonian rats as a viable model for testing aDBS based on beta power and provide a path to investigate more complex closed-loop algorithms in freely behaving animals.
Collapse
Affiliation(s)
- Judith Evers
- Neuromuscular Systems Lab, School of Electrical and Electronic Engineering, University College Dublin Belfield, Belfield, Dublin, Ireland.
| | - Jakub Orłowski
- Neuromuscular Systems Lab, School of Electrical and Electronic Engineering, University College Dublin Belfield, Belfield, Dublin, Ireland
| | - Hanne Jahns
- Department of Pathology, School of Veterinary Medicine, University College Dublin Belfield, Dublin, Ireland
| | - Madeleine M Lowery
- Neuromuscular Systems Lab, School of Electrical and Electronic Engineering, University College Dublin Belfield, Belfield, Dublin, Ireland
| |
Collapse
|
4
|
Pogorelov VM, Martini ML, Jin J, Wetsel WC, Caron MG. Dopamine-Depleted Dopamine Transporter Knockout (DDD) Mice: Dyskinesia with L-DOPA and Dopamine D1 Agonists. Biomolecules 2023; 13:1658. [PMID: 38002340 PMCID: PMC10669682 DOI: 10.3390/biom13111658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
L-DOPA is the mainstay of treatment for Parkinson's disease (PD). However, over time this drug can produce dyskinesia. A useful acute PD model for screening novel compounds for anti-parkinsonian and L-DOPA-induced dyskinesia (LID) are dopamine-depleted dopamine-transporter KO (DDD) mice. Treatment with α-methyl-para-tyrosine rapidly depletes their brain stores of DA and renders them akinetic. During sensitization in the open field (OF), their locomotion declines as vertical activities increase and upon encountering a wall they stand on one leg or tail and engage in climbing behavior termed "three-paw dyskinesia". We have hypothesized that L-DOPA induces a stereotypic activation of locomotion in DDD mice, where they are unable to alter the course of their locomotion, and upon encountering walls engage in "three-paw dyskinesia" as reflected in vertical counts or beam-breaks. The purpose of our studies was to identify a valid index of LID in DDD mice that met three criteria: (a) sensitization with repeated L-DOPA administration, (b) insensitivity to a change in the test context, and (c) stimulatory or inhibitory responses to dopamine D1 receptor agonists (5 mg/kg SKF81297; 5 and 10 mg/kg MLM55-38, a novel compound) and amantadine (45 mg/kg), respectively. Responses were compared between the OF and a circular maze (CM) that did not hinder locomotion. We found vertical counts and climbing were specific for testing in the OF, while oral stereotypies were sensitized to L-DOPA in both the OF and CM and responded to D1R agonists and amantadine. Hence, in DDD mice oral stereotypies should be used as an index of LID in screening compounds for PD.
Collapse
Affiliation(s)
- Vladimir M. Pogorelov
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, 354 Sands Building, 303 Research Drive, Durham, NC 27710, USA
| | - Michael L. Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.L.M.); (J.J.)
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.L.M.); (J.J.)
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, 354 Sands Building, 303 Research Drive, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA;
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Marc G. Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA;
| |
Collapse
|
5
|
Collier TJ, Begg L, Stancati JA, Mercado NM, Sellnow RC, Sandoval IM, Sortwell CE, Steece-Collier K. Quinpirole inhibits levodopa-induced dyskinesias at structural and behavioral levels: Efficacy negated by co-administration of isradipine. Exp Neurol 2023; 369:114522. [PMID: 37640098 PMCID: PMC10591902 DOI: 10.1016/j.expneurol.2023.114522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/06/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
Dopamine depletion associated with parkinsonism induces plastic changes in striatal medium spiny neurons (MSN) that are maladaptive and associated with the emergence of the negative side-effect of standard treatment: the abnormal involuntary movements termed levodopa-induced dyskinesia (LID). Prevention of MSN dendritic spine loss is hypothesized to diminish liability for LID in Parkinson's disease. Blockade of striatal CaV1.3 calcium channels can prevent spine loss and significantly diminish LID in parkinsonian rats. While pharmacological antagonism with FDA approved CaV1 L-type channel antagonist dihydropyridine (DHP) drugs (e.g, isradipine) are potentially antidyskinetic, pharmacologic limitations of current drugs may result in suboptimal efficacy. To provide optimal CaV1.3 antagonism, we investigated the ability of a dual pharmacological approach to more potently antagonize these channels. Specifically, quinpirole, a D2/D3-type dopamine receptor (D2/3R) agonist, has been demonstrated to significantly reduce calcium current activity at CaV1.3 channels in MSNs of rats by a mechanism distinct from DHPs. We hypothesized that dual inhibition of striatal CaV1.3 channels using the DHP drug isradipine combined with the D2/D3 dopamine receptor agonist quinpirole prior to, and in conjunction with, levodopa would be more effective at preventing structural modifications of dendritic spines and providing more stable LID prevention. For these proof-of-principle studies, rats with unilateral nigrostriatal lesions received daily administration of vehicle, isradipine, quinpirole, or isradipine + quinpirole prior to, and concurrent with, levodopa. Development of LID and morphological analysis of dendritic spines were assessed. Contrary to our hypothesis, quinpirole monotherapy was the most effective at reducing dyskinesia severity and preventing abnormal mushroom spine formation on MSNs, a structural phenomenon previously associated with LID. Notably, the antidyskinetic efficacy of quinpirole monotherapy was lost in the presence of isradipine co-treatment. These findings suggest that D2/D3 dopamine receptor agonists when given in combination with levodopa and initiated in early-stage Parkinson's disease may provide long-term protection against LID. The negative interaction of isradipine with quinpirole suggests a potential cautionary note for co-administration of these drugs in a clinical setting.
Collapse
Affiliation(s)
- Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, 220 Cherry St. S.E., Grand Rapids, MI 49503, USA.
| | - Lauren Begg
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Dr., Allendale, MI 49401, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA
| | - Rhyomi C Sellnow
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA; Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| | - Ivette M Sandoval
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, 220 Cherry St. S.E., Grand Rapids, MI 49503, USA.
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, 220 Cherry St. S.E., Grand Rapids, MI 49503, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave. N.W., Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, 220 Cherry St. S.E., Grand Rapids, MI 49503, USA
| |
Collapse
|
6
|
Han B, Wang M, Li J, Chen Q, Sun N, Yang X, Zhang Q. Perspectives and new aspects of histone deacetylase inhibitors in the therapy of CNS diseases. Eur J Med Chem 2023; 258:115613. [PMID: 37399711 DOI: 10.1016/j.ejmech.2023.115613] [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] [Received: 05/15/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/05/2023]
Abstract
Many populations worldwide are suffering from central nervous system (CNS) diseases such as brain tumors, neurodegenerative diseases (Alzheimer's disease, Parkinson's disease and Huntington's disease) and stroke. There is a shortage of effective drugs for most CNS diseases. As one of the regulatory mechanisms of epigenetics, the particular role and therapeutic benefits of histone deacetylases (HDACs) in the CNS have been extensively studied. In recent years, HDACs have attracted increasing attention as potential drug targets for CNS diseases. In this review, we summarize the recent applications of representative histone deacetylases inhibitors (HDACis) in CNS diseases and discuss the challenges in developing HDACis with different structures and better blood-brain barrier (BBB) permeability, hoping to promote the development of more effective bioactive HDACis for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Bo Han
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Mengfei Wang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Jiayi Li
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China; School of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Qiushi Chen
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China; School of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Niubing Sun
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China; School of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Xuezhi Yang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qingwei Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China.
| |
Collapse
|
7
|
Avila-Luna A, Gálvez-Rosas A, Aguirre-Pérez A, Hidalgo-Bravo A, Alfaro-Rodriguez A, Ríos C, Arias-Montaño JA, Bueno-Nava A. Chronic H 3R activation reduces L-Dopa-induced dyskinesia, normalizes cortical GABA and glutamate levels, and increases striatal dopamine D 1R mRNA expression in 6-hydroxydopamine-lesioned male rats. Psychopharmacology (Berl) 2023; 240:1221-1234. [PMID: 37086286 DOI: 10.1007/s00213-023-06339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/09/2023] [Indexed: 04/23/2023]
Abstract
RATIONALE Dyskinesias induced by L-3,4-dihydroxyphenylalanine, L-Dopa (LIDs), are the major complication in the pharmacological treatment of Parkinson's disease. LIDs induce overactivity of the glutamatergic cortico-striatal projections, and drugs that reduce glutamatergic overactivity exert antidyskinetic actions. Chronic administration of immepip, agonist at histamine H3 receptors (H3R), reduces LIDs and diminishes GABA and glutamate content in striatal dialysates (Avila-Luna et al., Psychopharmacology 236: 1937-1948, 2019). OBJECTIVES AND METHODS In rats unilaterally lesioned with 6-hydroxydopamine in the substantia nigra pars compacta (SNc), we examined whether the chronic administration of immepip and their withdrawal modify LIDs, the effect of L-Dopa on glutamate and GABA content, and mRNA levels of dopamine D1 receptors (D1Rs) and H3Rs in the cerebral cortex and striatum. RESULTS The administration of L-Dopa for 21 days induced LIDs. This effect was accompanied by increased GABA and glutamate levels in the cerebral cortex ipsi and contralateral to the lesioned SNc, and immepip administration prevented (GABA) or reduced (glutamate) these actions. In the striatum, GABA content increased in the ipsilateral nucleus, an effect prevented by immepip. L-Dopa administration had no significant effects on striatal glutamate levels. In lesioned and L-Dopa-treated animals, D1R mRNA decreased in the ipsilateral striatum, an effect prevented by immepip administration. CONCLUSIONS Our results indicate that chronic H3R activation reduces LIDs and the overactivity of glutamatergic cortico-striatal projections, providing further evidence for an interaction between D1Rs and H3Rs in the cortex and striatum under normal and pathological conditions.
Collapse
Affiliation(s)
- Alberto Avila-Luna
- Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
- Laboratorio de Neurofisiología Química de la Discapacidad, Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
| | - Arturo Gálvez-Rosas
- Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
- Laboratorio de Neurofisiología Química de la Discapacidad, Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
| | - Alexander Aguirre-Pérez
- Laboratorio de Neurofisiología Química de la Discapacidad, Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
| | - Alberto Hidalgo-Bravo
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
| | - Alfonso Alfaro-Rodriguez
- Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
| | - Camilo Ríos
- Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSa, Insurgentes Sur 3877, La Fama, Ciudad de México, 14269, México
- Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Calzada del Hueso 1100, Col. Villa Quietud, Ciudad de México, 04960, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN 2508, Zacatenco, Ciudad de México, 07360, México
| | - Antonio Bueno-Nava
- Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México.
- Laboratorio de Neurofisiología Química de la Discapacidad, Coordinación de Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, 14389, México.
| |
Collapse
|
8
|
Su D, Gan Y, Zhang Z, Cui Y, Zhang Z, Liu Z, Wang Z, Zhou J, Sossi V, Stoessl AJ, Wu T, Jing J, Feng T. Multimodal Imaging of Substantia Nigra in Parkinson's Disease with Levodopa-Induced Dyskinesia. Mov Disord 2023; 38:616-625. [PMID: 36799459 DOI: 10.1002/mds.29320] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/01/2022] [Accepted: 12/20/2022] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Degeneration of the substantia nigra (SN) may contribute to levodopa-induced dyskinesia (LID) in Parkinson's disease (PD), but the exact characteristics of SN in LID remain unclear. OBJECTIVE To further understand the pathogenesis of patients with PD with LID (PD-LID), we explored the structural and functional characteristics of SN in PD-LID using multimodal magnetic resonance imaging (MRI). METHODS Twenty-nine patients with PD-LID, 37 patients with PD without LID (PD-nLID), and 28 healthy control subjects underwent T1-weighted MRI, quantitative susceptibility mapping, neuromelanin-sensitive MRI, multishell diffusion MRI, and resting-state functional MRI. Different measures characterizing the SN were obtained using a region of interest-based approach. RESULTS Compared with patients with PD-nLID and healthy control subjects, the quantitative susceptibility mapping values of SN pars compacta (SNpc) were significantly higher (P = 0.049 and P = 0.00002), and the neuromelanin contrast-to-noise ratio values in SNpc were significantly lower (P = 0.012 and P = 0.000002) in PD-LID. The intracellular volume fraction of the posterior SN in PD-LID was significantly higher compared with PD-nLID (P = 0.037). Resting-state fMRI indicated that PD-LID in the medication off state showed higher functional connectivity between the SNpc and putamen compared with PD-nLID (P = 0.031), and the functional connectivity changes in PD-LID were positively correlated with Unified Dyskinesia Rating Scale total scores (R = 0.427, P = 0.042). CONCLUSIONS Our multimodal imaging findings highlight greater neurodegeneration in SN and the altered nigrostriatal connectivity in PD-LID. These characteristics provide a new perspective into the role of SN in the pathophysiological mechanisms underlying PD-LID. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dongning Su
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yawen Gan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhe Zhang
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,Tiantan Neuroimaging Center of Excellence, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yusha Cui
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhijin Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhu Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junhong Zhou
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Roslindale, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - A Jon Stoessl
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada.,Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tao Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Tiantan Neuroimaging Center of Excellence, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
9
|
Bourque M, Grégoire L, Patel W, Dickens D, Snodgrass R, Di Paolo T. AV-101, a Pro-Drug Antagonist at the NMDA Receptor Glycine Site, Reduces L-Dopa Induced Dyskinesias in MPTP Monkeys. Cells 2022; 11:cells11223530. [PMID: 36428960 PMCID: PMC9688762 DOI: 10.3390/cells11223530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors have been implicated in L-Dopa-induced dyskinesias (LID) in Parkinson's disease patients, but the use of antagonists that directly inhibit this receptor is associated with severe side effects. L-4-chlorokynurenine (4-Cl-KYN or AV-101) is a pro-drug of 7-chlorokynurenic acid (7-Cl-KYNA), a potent and specific antagonist of the glycine (GlyB) co-agonist site of NMDA receptors. The 7-Cl-KYNA has limited ability to cross the blood-brain barrier, whereas AV-101 readily accesses the brain. We investigated if AV-101 reduces LID in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys while maintaining the antiparkinsonian activity of L-Dopa. A first pilot study using three dyskinetic MPTP monkeys showed that acute AV-101 treatment (250 and 450 mg/kg) reduced LID and maintained the antiparkinsonian activity of L-Dopa. The main study using six additional dyskinetic MPTP monkeys showed that repeated AV-101 treatment (250 mg/kg, b.i.d. for 4 consecutive days) maintained their L-Dopa antiparkinsonian response. We measured significantly less LID when AV-101 was combined with L-Dopa treatment. AV-101 alone or with L-Dopa had no non-motor adverse effects in MPTP monkeys. Our study showed antidyskinetic activity of AV-101 in MPTP monkeys was comparable to amantadine tested previously in our laboratory in this model. We observed no adverse effects with AV-101, which is an improvement over amantadine, with its known side effects.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | - Laurent Grégoire
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
| | - Waseema Patel
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - David Dickens
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - Ralph Snodgrass
- Vistagen Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC G1V4G2, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC G1V0A6, Canada
- Correspondence:
| |
Collapse
|
10
|
Rota S, Urso D, van Wamelen DJ, Leta V, Boura I, Odin P, Espay AJ, Jenner P, Chaudhuri KR. Why do 'OFF' periods still occur during continuous drug delivery in Parkinson's disease? Transl Neurodegener 2022; 11:43. [PMID: 36229860 PMCID: PMC9558383 DOI: 10.1186/s40035-022-00317-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Continuous drug delivery (CDD) is used in moderately advanced and late-stage Parkinson’s disease (PD) to control motor and non-motor fluctuations (‘OFF’ periods). Transdermal rotigotine is indicated for early fluctuations, while subcutaneous apomorphine infusion and levodopa-carbidopa intestinal gel are utilised in advanced PD. All three strategies are considered examples of continuous dopaminergic stimulation achieved through CDD. A central premise of the CDD is to achieve stable control of the parkinsonian motor and non-motor states and avoid emergence of ‘OFF’ periods. However, data suggest that despite their efficacy in reducing the number and duration of ‘OFF’ periods, these strategies still do not prevent ‘OFF’ periods in the middle to late stages of PD, thus contradicting the widely held concepts of continuous drug delivery and continuous dopaminergic stimulation. Why these emergent ‘OFF’ periods still occur is unknown. In this review, we analyse the potential reasons for their persistence. The contribution of drug- and device-related involvement, and the problems related to site-specific drug delivery are analysed. We propose that changes in dopaminergic and non-dopaminergic mechanisms in the basal ganglia might render these persistent ‘OFF’ periods unresponsive to dopaminergic therapy delivered via CDD.
Collapse
Affiliation(s)
- Silvia Rota
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK. .,Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK. .,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Daniele Urso
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro, "Pia Fondazione Cardinale G. Panico", 73039, Tricase, Italy
| | - Daniel J van Wamelen
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Valentina Leta
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK
| | - Iro Boura
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,School of Medicine, University of Crete, Crete, Greece.,Department of Neurology, University Hospital of Heraklion, Crete, Greece
| | - Per Odin
- Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Alberto J Espay
- University of Cincinnati Gardner Neuroscience Institute, Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Jenner
- Institute of Pharmaceutical Sciences, Faculty of Life Science and Medicine, King's College London, London, UK.
| | - K Ray Chaudhuri
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK
| |
Collapse
|
11
|
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: 1.3] [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
|
12
|
Zhang Z, Li H, Su Y, Ma J, Yuan Y, Yu Z, Shi M, Shao S, Zhang Z, Hölscher C. Neuroprotective Effects of a Cholecystokinin Analogue in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Parkinson’s Disease Mouse Model. Front Neurosci 2022; 16:814430. [PMID: 35368248 PMCID: PMC8964967 DOI: 10.3389/fnins.2022.814430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 02/11/2022] [Indexed: 01/13/2023] Open
Abstract
Parkinson’s disease (PD) is a chronic neurodegenerative disease. Type 2 diabetes mellitus (T2DM) has been identified as a risk factor for PD. Drugs originally developed for T2DM treatment such as liraglutide have shown neuroprotective effects in mouse models of PD. Cholecystokinin (CCK) is a peptide hormone with growth factor properties. Here, we demonstrate the neuroprotective effects of the (pGLu)-(Gln)-CCK8 analogue in an acute PD mouse model induced by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Administration of CCK analogue (50 nmol/kg ip.) for 14 days treatment improved the locomotor and exploratory activity of mice, and improved bradykinesia and movement balance of mice. The CCK analogue administration also restored tyrosine hydroxylase (TH) positive dopaminergic neurons number and synapse number (synaptophysin levels) in the substantia nigra pars compacta (SNpc). The CCK analogue decreased glia activation and neuroinflammation in the SNpc, and regulated autophagy dysfunction induced by MPTP. CCK analogue protected against mitochondrial damage and ER stress, and also decreased the ratio of apoptosis signaling molecules Bax/Bcl-2. Importantly, the CCK analogue improved the decrease of p-CREBS133 growth factor signaling in the SNpc. Therefore, the CCK analogue promotes cell survival of dopaminergic neuron in the SNpc by activating the cAMP/PKA/CREB pathway that also inhibits apoptosis and regulates autophagy impairment. The present results indicate that CCK analogue shows a promising potential for the treatment of PD.
Collapse
Affiliation(s)
- Zijuan Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hai Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunfang Su
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinlian Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ye Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ziyang Yu
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ming Shi
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Simai Shao
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Zhenqiang Zhang,
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Neurology Department of the Second Associated Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Christian Hölscher,
| |
Collapse
|
13
|
Animal models of action control and cognitive dysfunction in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:227-255. [PMID: 35248196 DOI: 10.1016/bs.pbr.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) has historically been considered a motor disorder induced by a loss of dopaminergic neurons in the substantia nigra pars compacta. More recently, it has been recognized to have significant non-motor symptoms, most prominently cognitive symptoms associated with a dysexecutive syndrome. It is common in the literature to see motor and cognitive symptoms treated separately and, indeed, there has been a general call for specialized treatment of the latter, particularly in the more severe cases of PD with mild cognitive impairment and dementia. Animal studies have similarly been developed to model the motor or non-motor symptoms. Nevertheless, considerable research has established that segregating consideration of cognition from the precursors to motor movement, particularly movement associated with goal-directed action, is difficult if not impossible. Indeed, on some contemporary views cognition is embodied in action control, something that is particularly prevalent in theory and evidence relating to the integration of goal-directed and habitual control processes. The current paper addresses these issues within the literature detailing animal models of cognitive dysfunction in PD and their neural and neurochemical bases. Generally, studies using animal models of PD provide some of the clearest evidence for the integration of these action control processes at multiple levels of analysis and imply that consideration of this integrative process may have significant benefits for developing new approaches to the treatment of PD.
Collapse
|
14
|
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
|
15
|
Kumar V, Kundu S, Singh A, Singh S. Understanding the role of histone deacetylase and their inhibitors in neurodegenerative disorders: Current targets and future perspective. Curr Neuropharmacol 2021; 20:158-178. [PMID: 34151764 PMCID: PMC9199543 DOI: 10.2174/1570159x19666210609160017] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 11/28/2022] Open
Abstract
Neurodegenerative diseases are a group of pathological conditions that cause motor inc-ordination (jerking movements), cognitive and memory impairments result from degeneration of neurons in a specific area of the brain. Oxidative stress, mitochondrial dysfunction, excitotoxicity, neuroinflammation, neurochemical imbalance and histone deacetylase enzymes (HDAC) are known to play a crucial role in neurodegeneration. HDAC is classified into four categories (class I, II, III and class IV) depending upon their location and functions. HDAC1 and 2 are involved in neurodegeneration, while HDAC3-11 and class III HDACs are beneficial as neuroprotective. HDACs are localized in different parts of the brain- HDAC1 (hippocampus and cortex), HDAC2 (nucleus), HDAC3, 4, 5, 7 and 9 (nucleus and cytoplasm), HDAC6 & HDAC7 (cytoplasm) and HDAC11 (Nucleus, cornus ammonis 1 and spinal cord). In pathological conditions, HDAC up-regulates glutamate, phosphorylation of tau, and glial fibrillary acidic proteins while down-regulating BDNF, Heat shock protein 70 and Gelsolin. Class III HDACs are divided into seven sub-classes (SIRT1-SIRT7). Sirtuins are localized in the different parts of the brain and neuron -Sirt1 (nucleus), Sirt2 (cortex, striatum, hippocampus and spinal cord), Sirt3 (mitochondria and cytoplasm), Sirt4, Sirt5 & Sirt6 (mitochondria), Sirt7 (nucleus) and Sirt8 (nucleolus). SIRTs (1, 3, 4, and 6) are involved in neuronal survival, proliferation and modulating stress response, and SIRT2 is associated with Parkinsonism, Huntington’s disease and Alzheimer’s disease, whereas SIRT6 is only associated with Alzheimer’s disease. In this critical review, we have discussed the mechanisms and therapeutic targets of HDACs that would be beneficial for the management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Vishal Kumar
- Scholar, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Satyabrata Kundu
- Scholar, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Arti Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
16
|
Correlación entre la expresión del receptor dopaminérgico D2 y presencia de movimientos involuntarios anormales (MIA) en un modelo de disquinesia en ratas Wistar hemiparkinsonizadas. Neurologia 2021. [DOI: 10.1016/j.nrl.2017.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
17
|
Caro Aponte P, Otálora C, Guzmán J, Turner L, Alcázar J, Mayorga E. Correlation between dopamine receptor D2 expression and presence of abnormal involuntary movements in Wistar rats with hemiparkinsonism and dyskinesia. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2017.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
18
|
He YB, Liu YL, Yang ZD, Lu JH, Song Y, Guan YM, Chen YM. Effect of ginsenoside-Rg1 on experimental Parkinson's disease: A systematic review and meta-analysis of animal studies. Exp Ther Med 2021; 21:552. [PMID: 33850524 PMCID: PMC8027743 DOI: 10.3892/etm.2021.9984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 02/02/2021] [Indexed: 11/06/2022] Open
Abstract
Previous studies have reported that ginsenoside-Rg1 (G-Rg1) was able to mitigate the loss of dopaminergic neurons in animal models of Parkinson's disease (PD). The present study provided a systematic review and meta-analysis of preclinical studies to pool current evidence on the effect of G-Rg1 on neurogenesis in the treatment of PD. Eligible studies were identified through a search from six databases: PubMed, EMBASE, Web of Science, VIP, Chinese National Knowledge Infrastructure and the Wanfang database. Primary outcomes were tyrosine hydroxylase (TH)-positive cells in the nigra, Nissl staining-positive cells in the nigra, pole test time and dopamine (DA) levels in the striatum. A total of 18 eligible studies were identified, involving 343 animals. Of these, 13 reported a significant relationship between G-Rg1 and improved TH-positive cells in the nigra compared with the control group (P<0.00001). Furthermore, 3 studies reported a significant relationship between G-Rg1 and improved Nissl-positive cells in the nigra compared with the control group (P<0.00001). In addition, 4 studies reported a significant effect of G-Rg1 to reduce the total pole test time compared with that in the control group (P=0.001). A total of 3 studies indicated a significant association between G-Rg1 and improved DA levels in the striatum compared with the control group (P<0.00001). These results suggested that G-Rg1 has positive effects in attenuating damage in models of PD, and thus, it is a potential candidate neuroprotective drug for human PD.
Collapse
Affiliation(s)
- Yi-Bo He
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yong-Lin Liu
- Reproductive Center, Sanya Maternal and Child Health Center, Sanya, Hainan 572000, P.R. China
| | - Zheng-Dong Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Jia-Hong Lu
- Department of Obstetrics and Gynecology, The First People's Hospital of Xiaoshan, Hangzhou, Zhejiang 311200, P.R. China
| | - Yao Song
- Department of Acupuncture, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310015, P.R. China
| | - Yan-Ming Guan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yi-Min Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
19
|
The BDNF Val66Met polymorphism (rs6265) enhances dopamine neuron graft efficacy and side-effect liability in rs6265 knock-in rats. Neurobiol Dis 2020; 148:105175. [PMID: 33188920 PMCID: PMC7855552 DOI: 10.1016/j.nbd.2020.105175] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 01/10/2023] Open
Abstract
Prevalent in approximately 20% of the worldwide human population, the
rs6265 (also called ‘Val66Met’) single nucleotide polymorphism
(SNP) in the gene for brain-derived neurotrophic factor (BDNF)
is a common genetic variant that can alter therapeutic responses in individuals
with Parkinson’s disease (PD). Possession of the variant Met allele
results in decreased activity-dependent release of BDNF. Given the resurgent
worldwide interest in neural transplantation for PD and the biological relevance
of BDNF, the current studies examined the effects of the rs6265 SNP on
therapeutic efficacy and side-effect development following primary dopamine (DA)
neuron transplantation. Considering the significant reduction in BDNF release
associated with rs6265, we hypothesized that rs6265-mediated dysfunctional BDNF
signaling contributes to the limited clinical benefit observed in a
subpopulation of PD patients despite robust survival of grafted DA neurons, and
further, that this mutation contributes to the development of aberrant
graft-induced dyskinesias (GID). To this end, we generated a CRISPR knock-in rat
model of the rs6265 BDNF SNP to examine for the first time the
influence of a common genetic polymorphism on graft survival, functional
efficacy, and side-effect liability, comparing these parameters between
wild-type (Val/Val) rats and those homozygous for the variant Met allele
(Met/Met). Counter to our hypothesis, the current research indicates that
Met/Met rats show enhanced graft-associated therapeutic efficacy and a
paradoxical enhancement of graft-derived neurite outgrowth compared to wild-type
rats. However, consistent with our hypothesis, we demonstrate that the rs6265
genotype in the host rat is strongly linked to development of GID, and that this
behavioral phenotype is significantly correlated with neurochemical signatures
of atypical glutamatergic neurotransmission by grafted DA neurons.
Collapse
|
20
|
Lecours C, St-Pierre MK, Picard K, Bordeleau M, Bourque M, Awogbindin IO, Benadjal A, Ibanez FG, Gagnon D, Cantin L, Parent M, Di Paolo T, Tremblay ME. Levodopa partially rescues microglial numerical, morphological, and phagolysosomal alterations in a monkey model of Parkinson's disease. Brain Behav Immun 2020; 90:81-96. [PMID: 32755645 DOI: 10.1016/j.bbi.2020.07.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder. The mechanisms underlying the onset and progression of Levodopa (L-Dopa)-induced dyskinesia (LID) during PD treatment remain elusive. Emerging evidence implicates functional modification of microglia in the development of LID. Thus, understanding the link between microglia and the development of LID may provide the knowledge required to preserve or promote beneficial microglial functions, even during a prolonged L-Dopa treatment. To provide novel insights into microglial functional alterations in PD pathophysiology, we characterized their density, morphology, ultrastructure, and degradation activity in the sensorimotor functional territory of the putamen, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) cynomolgus monkeys. A subset of MPTP monkeys was treated orally with L-Dopa and developed LID similar to PD patients. Using a combination of light, confocal and transmission electron microscopy, our quantitative analyses revealed alterations of microglial density, morphology and phagolysosomal activity following MPTP intoxication that were partially normalized with L-Dopa treatment. In particular, microglial density, cell body and arborization areas were increased in the MPTP monkeys, whereas L-Dopa-treated MPTP animals presented a microglial phenotype similar to the control animals. At the ultrastructural level, microglia did not differ between groups in their markers of cellular stress or aging. Nevertheless, microglia from the MPTP monkeys displayed reduced numbers of endosomes, compared with control animals, that remained lower after L-Dopa treatment. Microglia from MPTP monkeys treated with L-Dopa also had increased numbers of primary lysosomes compared with non-treated MPTP animals, while secondary and tertiary lysosomes remained unchanged. Moreover, a decrease microglial immunoreactivity for CD68, considered a marker of phagocytosis and lysosomal activity, was measured in the MPTP monkeys treated with L-Dopa, compared with non-treated MPTP animals. Taken together, these findings revealed significant changes in microglia during PD pathophysiology that were partially rescued by L-Dopa treatment. Albeit, this L-Dopa treatment conferred phagolysosomal insufficiency on microglia in the dyskinetic Parkinsonian monkeys.
Collapse
Affiliation(s)
- Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Maude Bordeleau
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Integrated Program of Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Melanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Ifeoluwa Oluleke Awogbindin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Amin Benadjal
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Biologie Intégrative et Physiologie, Sorbonne Université, Paris VI, France
| | | | - Dave Gagnon
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada; CERVO Brain Research Center, Québec, QC, Canada
| | - Leo Cantin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada; CERVO Brain Research Center, Québec, QC, Canada
| | - Therese Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
21
|
Seo J, Won J, Kim K, Park J, Yeo HG, Kim YG, Baek SH, Lee H, Jeon CY, Choi WS, Lee S, Kim KJ, Park SH, Son Y, Jeong KJ, Lim KS, Kang P, Lee HY, Son HC, Huh JW, Kim YH, Lee DS, Lee SR, Choi JW, Lee Y. Impaired Hand Dexterity Function in a Non-human Primate Model with Chronic Parkinson's Disease. Exp Neurobiol 2020; 29:376-388. [PMID: 33154199 PMCID: PMC7649085 DOI: 10.5607/en20040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Symptoms of Parkinson’s disease (PD) caused by loss of dopaminergic neurons are accompanied by movement disorders, including tremors, rigidity, bradykinesia, and akinesia. Non-human primate (NHP) models with PD play an essential role in the analysis of PD pathophysiology and behavior symptoms. As impairments of hand dexterity function can affect activities of daily living in patients with PD, research on hand dexterity function in NHP models with chronic PD is essential. Traditional rating scales previously used in the evaluation of animal spontaneous behavior were insufficient due to factors related to subjectivity and passivity. Thus, experimentally designed applications for an appropriate apparatus are necessary. In this study, we aimed to longitudinally assess hand dexterity function using hand dexterity task (HDT) in NHP-PD models. To validate this assessment, we analyzed the alteration in Parkinsonian tremor signs and the functionality of presynaptic dopaminergic neuron using positron emission tomography imaging of dopamine transporters in these models. In addition, a significant inverse correlation between HDT and DAT level was identified, but no local bias was found. The correlation with intention tremor signs was lower than the resting tremor. In conclusion, the evaluation of HDT may reflect behavioral symptoms of NHP-PD models. Furthermore, HDT was effectively used to experimentally distinguish intention tremors from other tremors.
Collapse
Affiliation(s)
- Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Hyeon-Gu Yeo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Yu Gyeong Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Seung Ho Baek
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Hoonwon Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Sangil Lee
- Primate Resource Center, KRIBB, Jeongeup 56216, Korea
| | - Ki Jin Kim
- Primate Resource Center, KRIBB, Jeongeup 56216, Korea
| | - Sung-Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Yeonghoon Son
- Primate Resource Center, KRIBB, Jeongeup 56216, Korea
| | - Kang Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, KRIBB, Cheongju 28116, Korea
| | - Philyong Kang
- Futuristic Animal Resource & Research Center, KRIBB, Cheongju 28116, Korea
| | - Hwal-Yong Lee
- Futuristic Animal Resource & Research Center, KRIBB, Cheongju 28116, Korea
| | - Hee-Chang Son
- Futuristic Animal Resource & Research Center, KRIBB, Cheongju 28116, Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Ji-Woong Choi
- Brain Engineering Convergence Research Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea.,Department of Information and Communication Engineering, DGIST, Daegu 42988, Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
22
|
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: 9] [Impact Index Per Article: 1.8] [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
|
23
|
Garcia-Chica J, D Paraiso WK, Tanabe S, Serra D, Herrero L, Casals N, Garcia J, Ariza X, Quader S, Rodriguez-Rodriguez R. An overview of nanomedicines for neuron targeting. Nanomedicine (Lond) 2020; 15:1617-1636. [PMID: 32618490 DOI: 10.2217/nnm-2020-0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Medical treatments of neuron-related disorders are limited due to the difficulty of targeting brain cells. Major drawbacks are the presence of the blood-brain barrier and the lack of specificity of the drugs for the diseased cells. Nanomedicine-based approaches provide promising opportunities for overcoming these limitations. Although many previous reviews are focused on brain targeting with nanomedicines in general, none of those are concerned explicitly on the neurons, while targeting neuronal cells in central nervous diseases is now one of the biggest challenges in nanomedicine and neuroscience. We review the most relevant advances in nanomedicine design and strategies for neuronal drug delivery that might successfully bridge the gap between laboratory and bedside treatment in neurology.
Collapse
Affiliation(s)
- Jesus Garcia-Chica
- Department of Basic Sciences, Faculty of Medicine & Health Sciences, Universitat Internacional de Catalunya, 08195, Sant Cugat del Vallès, Spain
- Department of Biochemistry & Physiology, School of Pharmacy & Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
| | - West Kristian D Paraiso
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, 210-0821, Japan
| | - Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety & Research, National Institute of Health Sciences, Kawasaki, Kanagawa, 210-9501, Japan
| | - Dolors Serra
- Department of Biochemistry & Physiology, School of Pharmacy & Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry & Physiology, School of Pharmacy & Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Núria Casals
- Department of Basic Sciences, Faculty of Medicine & Health Sciences, Universitat Internacional de Catalunya, 08195, Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Jordi Garcia
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Xavier Ariza
- Department of Inorganic & Organic Chemistry, Faculty of Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Sabina Quader
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, 210-0821, Japan
| | - Rosalia Rodriguez-Rodriguez
- Department of Basic Sciences, Faculty of Medicine & Health Sciences, Universitat Internacional de Catalunya, 08195, Sant Cugat del Vallès, Spain
| |
Collapse
|
24
|
Harsanyiova J, Buday T, Kralova Trancikova A. Parkinson's Disease and the Gut: Future Perspectives for Early Diagnosis. Front Neurosci 2020; 14:626. [PMID: 32625058 PMCID: PMC7313629 DOI: 10.3389/fnins.2020.00626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive degeneration of dopaminergic neurons, and at the cellular level by the formation of Lewy bodies in the central nervous system (CNS). However, the onset of the disease is believed to be localized to peripheral organs, particularly the gastrointestinal tract (GIT) and the olfactory bulb sooner before neuropathological changes occur in the CNS. Patients already in the pre-motor stage of PD suffer from various digestive problems and/or due to significant changes in the composition of the intestinal microbiome in this early stage of the disease. Detailed analyses of patient biopsies and autopsies as well as animal models of neuropathological changes characteristic of PD provided important information on the pathology or treatment of PD symptoms. However, presently is not clarified (i) the specific tissue in the GIT where the pathological processes associated with PD is initiated; (ii) the mechanism by which these processes are disseminated to the CNS or other tissues within the GIT; and (iii) which neuropathological changes could also serve as a reliable diagnostic marker of the premotor stages of PD, or (iv) which type of GIT tissue would be the most appropriate choice for routine examination of patient biopsies.
Collapse
Affiliation(s)
- Jana Harsanyiova
- Departmet of Pahophysiology, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| | - Tomas Buday
- Departmet of Pahophysiology, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| | - Alzbeta Kralova Trancikova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| |
Collapse
|
25
|
Steece-Collier K, Collier TJ, Lipton JW, Stancati JA, Winn ME, Cole-Strauss A, Sellnow R, Conti MM, Mercado NM, Nillni EA, Sortwell CE, Manfredsson FP, Bishop C. Striatal Nurr1, but not FosB expression links a levodopa-induced dyskinesia phenotype to genotype in Fisher 344 vs. Lewis hemiparkinsonian rats. Exp Neurol 2020; 330:113327. [PMID: 32387398 DOI: 10.1016/j.expneurol.2020.113327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/23/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
Abstract
Numerous genes, and alterations in their expression, have been identified as risk factors for developing levodopa-induced dyskinesia (LID). However, our understanding of the complexities of molecular changes remains insufficient for development of clinical treatment. In the current study we used gene array, in situ hybridization, immunohistochemistry, and microdialysis to provide a unique compare and contrast assessment of the relationship of four candidate genes to LID, employing three genetically distinct rat strains (Sprague-Dawley (SD), Fischer-344 (F344) and Lewis-RT.1) showing differences in dyskinesia susceptibility and 'first-ever LID' versus 'chronic LID' expression in subjects displaying equal dyskinesia severity. In these studies, rat strains were easily distinguishable for their LID propensity with: 1) a majority of SD rats expressing LID (LID+) and a subset being resistant (LID-); 2) all F344 rats readily developing (LID+); and 3) all Lewis rats being LID-resistant (LID-). Following chronic levodopa, LID+ SD rats showed significant increases in candidate gene expression: Nr4a2/(Nurr1) > > Trh > Inhba = Fosb. However, SD rats with long-standing striatal dopamine (DA) depletion treated with first-ever versus chronic high-dose levodopa revealed that despite identical levels of LID severity: 1) Fosb and Nurr1 transcripts but not protein were elevated with acute LID expression; 2) FOSB/ΔFOSB and NURR1 proteins were elevated only with chronic LID; and 3) Trh transcript and protein were elevated only with chronic LID. Strikingly, despite similar levodopa-induced striatal DA release in both LID-expressing F344 and LID-resistant Lewis rats, Fosb, Trh, Inhba transcripts were significantly elevated in both strains; however, Nurr1 mRNA was significantly increased only in LID+ F344 rats. These findings suggest a need to reevaluate currently accepted genotype-to-phenotype relationships in the expression of LID, specifically that of Fosb, a transcription factor generally assumed to play a causal role, and Nurr1, a transcription factor that has received significant attention in PD research linked to its critical role in the survival and function of midbrain DA neurons but who's striatal expression, generally below levels of detection, has remained largely unexplored as a regulator of LID. Finally these studies introduce a novel 'model' (inbred F344 vs inbred Lewis) that may provide a powerful tool for investigating the role for 'dyskinesia-resistance' genes downstream of 'dyskinesia-susceptibility' genes in modulating LID expression, a concept that has received considerably less attention and offers a new ways of thinking about antidyskinetic therapies.
Collapse
Affiliation(s)
- Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA.
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - Jack W Lipton
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Mary E Winn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Rhyomi Sellnow
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Melissa M Conti
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Eduardo A Nillni
- Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, USA
| |
Collapse
|
26
|
Chen X, Wang Y, Wu H, Cheng C, Le W. Research advances on L-DOPA-induced dyskinesia: from animal models to human disease. Neurol Sci 2020; 41:2055-2065. [DOI: 10.1007/s10072-020-04333-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/07/2020] [Indexed: 02/06/2023]
|
27
|
Taguchi T, Ikuno M, Yamakado H, Takahashi R. Animal Model for Prodromal Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21061961. [PMID: 32183024 PMCID: PMC7139491 DOI: 10.3390/ijms21061961] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and subsequent motor symptoms, but various non-motor symptoms (NMS) often precede motor symptoms. Recently, NMS have attracted much attention as a clue for identifying patients in a prodromal stage of PD, which is an excellent point at which to administer disease-modifying therapies (DMTs). These prodromal symptoms include olfactory loss, constipation, and sleep disorders, especially rapid eye movement sleep behavior disorder (RBD), all of which are also important for elucidating the mechanisms of the initiation and progression of the disease. For the development of DMTs, an animal model that reproduces the prodromal stage of PD is also needed. There have been various mammalian models reported, including toxin-based, genetic, and alpha synuclein propagation models. In this article, we review the animal models that exhibit NMS as prodromal symptoms and also discuss an appropriate prodromal model and its importance for the development of DMT of PD.
Collapse
Affiliation(s)
| | | | - Hodaka Yamakado
- Correspondence: (H.Y.); (R.T.); Tel.: +81-75-751-3767 (H.Y.); Tel.: +81-75-751-4397 (R.T.); Fax: +81-75-761-9780 (H.Y.); Fax: +81-75-761-9780 (R.T.)
| | - Ryosuke Takahashi
- Correspondence: (H.Y.); (R.T.); Tel.: +81-75-751-3767 (H.Y.); Tel.: +81-75-751-4397 (R.T.); Fax: +81-75-761-9780 (H.Y.); Fax: +81-75-761-9780 (R.T.)
| |
Collapse
|
28
|
Maimaitiming A, Xiao K, Hu C, Chen J, Yang XH, Zhou DH, Gao LP, Dong XP, Shi Q. Aberrant Decrease of the Endogenous SIRT3 and Increases of Acetylated Proteins in Scrapie-Infected Cell Line SMB-S15 and in the Brains of Experimental Mice. ACS Chem Neurosci 2019; 10:4293-4302. [PMID: 31545894 DOI: 10.1021/acschemneuro.9b00341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The linkage between mitochondrial dysfunction and neurodegenerative diseases including prion diseases has been frequently reported. As the major deacetylase in mitochondria, SIRT3 plays a crucial part in regulating the function of many mitochondrial proteins. Although SIRT3 was reported to be linked to several neurodegenerative diseases, it is still unknown if SIRT3 is involved in prion diseases. In this study, we have presented a substantially declined status of mitochondrial SIRT3 in both the levels of cultured cells and an experimental rodent model during scrapie prion replication and infection. Such decreased SIRT3 activity led to a decreased deacetylating activity, resulting in increases of the acetylated forms of some substrates of SIRT3 in cells, such as SOD2 and ATP5β. Declined SOD2 and ATP5β activities subsequently caused an increase of intracellular ROS and a reduction of ATP. Furthermore, we have also proposed evidence that the activity of cellular SIRT3 is partially recovered when abnormal prion propagation in the cultured cells is removed by resveratrol. Those data emphasize a close connection between the prion replication and mitochondrial deacetylation due to SIRT3, thereby partially explaining mitochondrial dysfunction in prion diseases.
Collapse
Affiliation(s)
- Adalaiti Maimaitiming
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Chao Hu
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Jia Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Xue-Hua Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Dong-Hua Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Li-Ping Gao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
- Center for Global Public Health, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| |
Collapse
|
29
|
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: 1.5] [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
|
30
|
Gątarek P, Pawełczyk M, Jastrzębski K, Głąbiński A, Kałużna-Czaplińska J. Analytical methods used in the study of Parkinson's disease. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.05.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Steece‐Collier K, Stancati JA, Collier NJ, Sandoval IM, Mercado NM, Sortwell CE, Collier TJ, Manfredsson FP. Genetic silencing of striatal CaV1.3 prevents and ameliorates levodopa dyskinesia. Mov Disord 2019; 34:697-707. [PMID: 31002755 PMCID: PMC6563183 DOI: 10.1002/mds.27695] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Levodopa-induced dyskinesias are an often debilitating side effect of levodopa therapy in Parkinson's disease. Although up to 90% of individuals with PD develop this side effect, uniformly effective and well-tolerated antidyskinetic treatment remains a significant unmet need. The pathognomonic loss of striatal dopamine in PD results in dysregulation and disinhibition of striatal CaV1.3 calcium channels, leading to synaptopathology that appears to be involved in levodopa-induced dyskinesias. Although there are clinically available drugs that can inhibit CaV1.3 channels, they are not adequately potent and have only partial and transient impact on levodopa-induced dyskinesias. METHODS To provide unequivocal target validation, free of pharmacological limitations, we developed a CaV1.3 shRNA to provide high-potency, target-selective, mRNA-level silencing of striatal CaV1.3 channels and examined its ability to impact levodopa-induced dyskinesias in severely parkinsonian rats. RESULTS We demonstrate that vector-mediated silencing of striatal CaV1.3 expression in severely parkinsonian rats prior to the introduction of levodopa can uniformly and completely prevent induction of levodopa-induced dyskinesias, and this antidyskinetic benefit persists long term and with high-dose levodopa. In addition, this approach is capable of ameliorating preexisting severe levodopa-induced dyskinesias. Importantly, motoric responses to low-dose levodopa remained intact in the presence of striatal CaV1.3 silencing, indicating preservation of levodopa benefit without dyskinesia liability. DISCUSSION The current data provide some of the most profound antidyskinetic benefit reported to date and suggest that genetic silencing of striatal CaV1.3 channels has the potential to transform treatment of individuals with PD by allowing maintenance of motor benefit of levodopa in the absence of the debilitating levodopa-induced dyskinesia side effect. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kathy Steece‐Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Jennifer A. Stancati
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Nicholas J. Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Ivette M. Sandoval
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Natosha M. Mercado
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Caryl E. Sortwell
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Timothy J. Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Fredric P. Manfredsson
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| |
Collapse
|
32
|
Lafuente JV, Requejo C, Ugedo L. Nanodelivery of therapeutic agents in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2019; 245:263-279. [PMID: 30961870 DOI: 10.1016/bs.pbr.2019.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) as a motor disorder is pathologically featured by the loss of dopaminergic neurons of the substantia nigra compacta (SNc) and the consequent depletion of dopamine in the striatum. However, motor signs are detectable when the loss of dopaminergic striatal terminals exceeds to the dopaminergic neuronal degeneration in SN. Hence, recent evidences about the topological organization of the nigrostriatal system could provide novel insights about the progression of the neurodegenerative process as well as the correct application of the novel therapeutic strategies. Though dopaminergic drugs and different routes of administration have been proposed to treat PD, most of the effects are symptomatic with temporary effects resorting to invasive procedures to ameliorate the side effects. Since the blood-brain barrier (BBB) is the main obstacle for most of molecules to access to the brain, ongoing research is focused on halting the progression of PD through the use of those technologies that allow the effective delivery and diffusion of therapeutic molecules to the central nervous system for bypassing BBB and avoiding the side effects. In this context, nanotechnology is emerging as a promising tool for drug delivery. In fact, nanodelivery of restorative treatments in PD, such as gene therapy increased the effectiveness of neurotrophic factors for restoring the dopamine deficit and improving motor deficit in rodent models. Therefore, the present review is focused on the description and identification of the available nanotherapies developed in experimental models of PD which could suppose an important advance for controlled delivery of nanobioactive components into the brain and one more step for the clinical projection.
Collapse
Affiliation(s)
- José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Catalina Requejo
- Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Luisa Ugedo
- Neuropharmacology Group, University of the Basque Country (UPV-EHU), Leioa, Spain
| |
Collapse
|
33
|
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: 1.1] [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
|
34
|
Seo J, Lee Y, Kim BS, Park J, Yang S, Yoon HJ, Yoo J, Park HS, Hong JJ, Koo BS, Baek SH, Jeon CY, Huh JW, Kim YH, Park SJ, Won J, Ahn YJ, Kim K, Jeong KJ, Kang P, Lee DS, Lim SM, Jin YB, Lee SR. A non-human primate model for stable chronic Parkinson's disease induced by MPTP administration based on individual behavioral quantification. J Neurosci Methods 2018; 311:277-287. [PMID: 30391524 DOI: 10.1016/j.jneumeth.2018.10.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND The guidelines for applying individual adjustments to macaques according to the severity of behavioral symptoms during 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment were provided to reproduce stable chronic Parkinsonism in a recent study (Potts et al., 2014). But, since there are insufficient guidelines regarding objective severity criteria of individual symptoms for adjustments of MPTP treatment, it is difficult to develop MPTP-induced chronic non-human primate (NHP) models with stable symptoms. NEW METHOD The individual adjustments of MPTP administration based on results of automatic quantification of global activity (GA) using a video-based tracking system were applied to develop MPTP-PD model. Low-dose (0.2 mg/kg) intramuscular injection was repeated continuously until GA was lower than 8% of baseline Parkinsonian behavior scores. The positron emission tomography imaging were used to follow the longitudinal course of Parkinson's disease (PD). RESULTS Significant reductions in GA and dopamine transporter activity, along with significant increases in Parkinsonian behavior scores were found from 4 to 48 weeks following the first administration. GA was correlated with the Parkinsonian behavior score. The dopamine transporter activity was correlated with GA and the Parkinsonian behavior score. However, it was not correlated with the total dose of MPTP. Damage of dopaminergic neuronal systems in the basal ganglia was confirmed by immunohistochemistry and Western blot. COMPARISON WITH EXISTING METHOD This study reinforces previous guidelines regarding production of NHP models with stable Parkinsonian symptoms. CONCLUSIONS This novel strategy of MPTP administration based on global activity evaluations provides an important conceptual advance for the development of chronic NHP Parkinsonian models.
Collapse
Affiliation(s)
- Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Bom Sahn Kim
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Sejung Yang
- Department of Biomedical Engineering, Yonsei University, Wonju 220-710, Republic of Korea
| | - Hai-Jeon Yoon
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Jang Yoo
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Hyun Soo Park
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jung-Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Seung Ho Baek
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Sang Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Yu-Jin Ahn
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Physical Therapy, Graduate School of Inje University, Gimhae, Republic of Korea
| | - Kang Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Philyong Kang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Soo Mee Lim
- Department of Radiology, Ewha Womans University School of Medicine, Seoul, Republic of Korea.
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea.
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
35
|
Gardoni F, Morari M, Kulisevsky J, Brugnoli A, Novello S, Pisanò CA, Caccia C, Mellone M, Melloni E, Padoani G, Sosti V, Vailati S, Keywood C. Safinamide Modulates Striatal Glutamatergic Signaling in a Rat Model of Levodopa-Induced Dyskinesia. J Pharmacol Exp Ther 2018; 367:442-451. [DOI: 10.1124/jpet.118.251645] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 09/17/2018] [Indexed: 11/22/2022] Open
|
36
|
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: 2.3] [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]
|
37
|
Charvin D, Di Paolo T, Bezard E, Gregoire L, Takano A, Duvey G, Pioli E, Halldin C, Medori R, Conquet F. An mGlu4-Positive Allosteric Modulator Alleviates Parkinsonism in Primates. Mov Disord 2018; 33:1619-1631. [PMID: 30216534 DOI: 10.1002/mds.27462] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 04/19/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Levodopa remains the gold-standard treatment for PD. However, it becomes less effective as the disease progresses and produces debilitating side effects, such as motor fluctuations and l-dopa-induced dyskinesia. Modulation of metabotropic glutamate receptor 4 represents a promising antiparkinsonian approach in combination with l-dopa, but it has not been demonstrated in primates. OBJECTIVE We studied whether a novel positive allosteric modulator of the metabotropic glutamate receptor 4, PXT002331 (foliglurax), could reduce parkinsonism in primate models. METHODS We assessed the therapeutic potential of PXT002331 in three models of MPTP-induced parkinsonism in macaques. These models represent three different stages of disease evolution: early stage and advanced stage with and without l-dopa-induced dyskinesia. RESULTS As an adjunct to l-dopa, PXT002331 induced a robust and dose-dependent reversal of parkinsonian motor symptoms in macaques, including bradykinesia, tremor, posture, and mobility. Moreover, PXT002331 strongly decreased dyskinesia severity, thus having therapeutic efficacy on both parkinsonian motor impairment and l-dopa-induced dyskinesia. PXT002331 brain penetration was also assessed using PET imaging in macaques, and pharmacodynamic analyses support target engagement in the therapeutic effects of PXT002331. CONCLUSIONS This work provides a demonstration that a positive allosteric modulator of metabotropic glutamate receptor 4 can alleviate the motor symptoms of PD and the motor complications induced by l-dopa in primates. PXT002331 is the first compound of its class to enter phase IIa clinical trials. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Delphine Charvin
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Therese Di Paolo
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Erwan Bezard
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Laurent Gregoire
- Neuroscience Research Unit CHU de Québec, CHUL Pavillon and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Akihiro Takano
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Guillaume Duvey
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Elsa Pioli
- Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Christer Halldin
- Karolinska Institutet, Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Rossella Medori
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - François Conquet
- Prexton Therapeutics SA, 1228 Plan-les-Ouates, Geneva, Switzerland
| |
Collapse
|
38
|
Lecours C, Bordeleau M, Cantin L, Parent M, Paolo TD, Tremblay MÈ. Microglial Implication in Parkinson's Disease: Loss of Beneficial Physiological Roles or Gain of Inflammatory Functions? Front Cell Neurosci 2018; 12:282. [PMID: 30214398 PMCID: PMC6125334 DOI: 10.3389/fncel.2018.00282] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/09/2018] [Indexed: 01/12/2023] Open
Abstract
Microglia, often described as the brain-resident macrophages, play crucial roles in central nervous system development, maintenance, plasticity, and adaptation to the environment. Both aging and chronic stress promote microglial morphological and functional changes, which can lead to the development of brain pathologies including Parkinson's disease (PD). Indeed, aging, and chronic stress represent main environmental risk factors for PD. In these conditions, microglia are known to undergo different morphological and functional changes. Inflammation is an important component of PD and disequilibrium between pro- and anti-inflammatory microglial functions might constitute a crucial component of PD onset and progression. Cumulated data also suggest that, during PD, microglia might lose beneficial functions and gain detrimental ones, in addition to mediating inflammation. In this mini-review, we aim to summarize the literature discussing the functional and morphological changes that microglia undergo in PD pathophysiology and upon exposure to its two main environmental risk factors, aging, and chronic stress.
Collapse
Affiliation(s)
- Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Quebec, QC, Canada
| | - Maude Bordeleau
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Integrated Program of Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Léo Cantin
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Martin Parent
- CERVO Brain Research Centre, Quebec, QC, Canada
- Département de Psychiatrie et Neurosciences, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Quebec, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| |
Collapse
|
39
|
Antipova V, Wree A, Holzmann C, Mann T, Palomero-Gallagher N, Zilles K, Schmitt O, Hawlitschka A. Unilateral Botulinum Neurotoxin-A Injection into the Striatum of C57BL/6 Mice Leads to a Different Motor Behavior Compared with Rats. Toxins (Basel) 2018; 10:E295. [PMID: 30018211 PMCID: PMC6070800 DOI: 10.3390/toxins10070295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/13/2018] [Accepted: 07/15/2018] [Indexed: 12/26/2022] Open
Abstract
Different morphological changes in the caudate-putamen (CPu) of naïve rats and mice were observed after intrastriatal botulinum neurotoxin-A (BoNT-A) injection. For this purpose we here studied various motor behaviors in mice (n = 46) longitudinally up to 9 months after intrastriatal BoNT-A administration as previously reported for rats, and compared both outcomes. Apomorphine- and amphetamine-induced rotational behavior, spontaneous motor behavior, as well as lateralized neglect were studied in mice after the injection of single doses of BoNT-A into the right CPu, comparing them with sham-injected animals. Unilateral intrastriatal injection of BoNT-A in mice induced significantly increased contralateral apomorphine-induced rotations for 1 to 3 months, as well as significantly increased contralateral amphetamine-induced rotations 1 to 9 months after injection. In rats (n = 28), unilateral BoNT-A injection also induced significantly increased contralateral apomorphine-induced rotations 3 months after injection, but did not provoke amphetamine-induced rotations at all. Lateralized sensorimotor integration, forelimb preference, and forelimb stepping were significantly impaired on the left side. The differences in motor behaviors between rats and mice may be caused by different BoNT-A effects on cholinergic and catecholaminergic fibers in rat and mouse striata, interspecies differences in striatal receptor densities, and different connectomes of the basal ganglia.
Collapse
Affiliation(s)
- Veronica Antipova
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Macroscopic and Clinical Anatomy, Medical University of Graz, Harrachgasse 21/1, A-8010 Graz, Austria.
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, Ernst-Heydemann-Strasse 8, D-18057 Rostock, Germany.
| | - Teresa Mann
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
| | - Karl Zilles
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
- JARA-Translational Brain Medicine, D-52062 Aachen, Germany.
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Alexander Hawlitschka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| |
Collapse
|
40
|
Johnston TH, Lacoste AMB, Visanji NP, Lang AE, Fox SH, Brotchie JM. Repurposing drugs to treat l-DOPA-induced dyskinesia in Parkinson's disease. Neuropharmacology 2018; 147:11-27. [PMID: 29907424 DOI: 10.1016/j.neuropharm.2018.05.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 01/05/2023]
Abstract
In this review, we discuss the opportunity for repurposing drugs for use in l-DOPA-induced dyskinesia (LID) in Parkinson's disease. LID is a particularly suitable indication for drug repurposing given its pharmacological diversity, translatability of animal-models, availability of Phase II proof-of-concept (PoC) methodologies and the indication-specific regulatory environment. A compound fit for repurposing is defined as one with appropriate human safety-data as well as animal safety, toxicology and pharmacokinetic data as found in an Investigational New Drug (IND) package for another indication. We first focus on how such repurposing candidates can be identified and then discuss development strategies that might progress such a candidate towards a Phase II clinical PoC. We discuss traditional means for identifying repurposing candidates and contrast these with newer approaches, especially focussing on the use of computational and artificial intelligence (AI) platforms. We discuss strategies that can be categorised broadly as: in vivo phenotypic screening in a hypothesis-free manner; in vivo phenotypic screening based on analogy to a related disorder; hypothesis-driven evaluation of candidates in vivo and in silico screening with a hypothesis-agnostic component to the selection. To highlight the power of AI approaches, we describe a case study using IBM Watson where a training set of compounds, with demonstrated ability to reduce LID, were employed to identify novel repurposing candidates. Using the approaches discussed, many diverse candidates for repurposing in LID, originally envisaged for other indications, will be described that have already been evaluated for efficacy in non-human primate models of LID and/or clinically. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.
Collapse
Affiliation(s)
- Tom H Johnston
- Krembil Research Institute, University Health Network, Toronto, ON, Canada; Atuka Inc., Toronto, ON, Canada.
| | | | - Naomi P Visanji
- Edmund J Safra Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto Western Hospital, Toronto, ON, Canada
| | - Anthony E Lang
- Edmund J Safra Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto Western Hospital, Toronto, ON, Canada
| | - Susan H Fox
- Edmund J Safra Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto Western Hospital, Toronto, ON, Canada
| | - Jonathan M Brotchie
- Krembil Research Institute, University Health Network, Toronto, ON, Canada; Atuka Inc., Toronto, ON, Canada
| |
Collapse
|
41
|
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: 0.4] [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
|
42
|
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: 7.3] [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
|
43
|
Lafuente JV, Requejo C, Carrasco A, Bengoetxea H. Nanoformulation: A Useful Therapeutic Strategy for Improving Neuroprotection and the Neurorestorative Potential in Experimental Models of Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 137:99-122. [PMID: 29132545 DOI: 10.1016/bs.irn.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disorder, but current therapies are only symptomatic. Experimental models are necessary to go deeper in the comprehension of pathophysiological mechanism and to assess new therapeutic strategies. The unilateral 6-hydroxydopamine (6-OHDA) lesion either in medial forebrain bundle (MFB) or into the striatum in rats affords to study various stages of PD depending on the evolution time lapsed. A promising alternative to address the neurodegenerative process is the use of neurotrophic factors; but its clinical use has been limited due to its short half-life and rapid degradation after in vivo administration, along with difficulties for crossing the blood-brain barrier (BBB). Tyrosine hydroxylase (TH) immunostaining revealed a significant decrease of the TH-immunopositive striatal volume in 6-OHDA group from rostral to caudal one. The loss of TH-ir neurons and axodendritic network (ADN) was higher in caudal sections showing a selective vulnerability of the topological distributed dopaminergic system. In addition to a remarkable depletion of dopamine in the nigrostriatal system, the administration of 6-OHDA into MFB induces some other neuropathological changes such as an increase of glial fibrillary acidic protein (GFAP) positive cells in substantia nigra (SN) as well as in striatum. Intrastriatal implantation of micro- or nanosystems delivering neurotrophic factor in parkinsonized rats for bypassing BBB leads to a significative functional and morphological recovery. Neurorestorative morphological changes (preservation of the TH-ir cells and ADN) along the rostrocaudal axis of caudoputamen complex and SN have been probed supporting a selective recovering after the treatment as well. Others innovative therapeutic strategies, such as the intranasal delivery, have been recently assessed in order to search the NTF effects. In addition some others methodological key points are reviewed.
Collapse
Affiliation(s)
- Jose V Lafuente
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.
| | - Catalina Requejo
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Alejandro Carrasco
- Group Nanoneurosurgery, Institute of Health Research Biocruces, Barakaldo, Spain; Service Neurosurgery, Cruces University Hospital, Barakaldo, Spain
| | - Harkaitz Bengoetxea
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| |
Collapse
|
44
|
Masilamoni GJ, Smith Y. Chronic MPTP administration regimen in monkeys: a model of dopaminergic and non-dopaminergic cell loss in Parkinson's disease. J Neural Transm (Vienna) 2017; 125:337-363. [PMID: 28861737 DOI: 10.1007/s00702-017-1774-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/29/2017] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder clinically characterized by cardinal motor deficits including bradykinesia, tremor, rigidity and postural instability. Over the past decades, it has become clear that PD symptoms extend far beyond motor signs to include cognitive, autonomic and psychiatric impairments, most likely resulting from cortical and subcortical lesions of non-dopaminergic systems. In addition to nigrostriatal dopaminergic degeneration, pathological examination of PD brains, indeed, reveals widespread distribution of intracytoplasmic inclusions (Lewy bodies) and death of non-dopaminergic neurons in the brainstem and thalamus. For that past three decades, the MPTP-treated monkey has been recognized as the gold standard PD model because it displays some of the key behavioral and pathophysiological changes seen in PD patients. However, a common criticism raised by some authors about this model, and other neurotoxin-based models of PD, is the lack of neuronal loss beyond the nigrostriatal dopaminergic system. In this review, we argue that this assumption is largely incorrect and solely based on data from monkeys intoxicated with acute administration of MPTP. Work achieved in our laboratory and others strongly suggest that long-term chronic administration of MPTP leads to brain pathology beyond the dopaminergic system that displays close similarities to that seen in PD patients. This review critically examines these data and suggests that the chronically MPTP-treated nonhuman primate model may be suitable to study the pathophysiology and therapeutics of some non-motor features of PD.
Collapse
Affiliation(s)
- Gunasingh J Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30322, USA.
- Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30322, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30322, USA
- Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30322, USA
| |
Collapse
|
45
|
Requejo C, Ruiz-Ortega JA, Cepeda H, Sharma A, Sharma HS, Ozkizilcik A, Tian R, Moessler H, Ugedo L, Lafuente JV. Nanodelivery of Cerebrolysin and Rearing in Enriched Environment Induce Neuroprotective Effects in a Preclinical Rat Model of Parkinson’s Disease. Mol Neurobiol 2017; 55:286-299. [DOI: 10.1007/s12035-017-0741-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
46
|
Involvement of the kynurenine pathway in the pathogenesis of Parkinson’s disease. Prog Neurobiol 2017; 155:76-95. [DOI: 10.1016/j.pneurobio.2015.12.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022]
|
47
|
Abstract
The loss of nigrostriatal dopamine (DA) is the primary cause of motor dysfunction in Parkinson's disease (PD), but the underlying striatal mechanisms remain unclear. In spite of abundant literature portraying structural, biochemical and plasticity changes of striatal projection neurons (SPNs), in the past there has been a data vacuum from the natural human disease and its close model in non-human primates. Recently, single-cell recordings in advanced parkinsonian primates have generated new insights into the altered function of SPNs. Currently, there are also human data that provide direct evidence of profoundly dysregulated SPN activity in PD. Here, we review primate recordings that are impacting our understanding of the striatal dysfunction after DA loss, particularly through the analysis of physiologic correlates of parkinsonian motor behaviors. In contrast to recordings in rodents, data obtained in primates and patients demonstrate similar major abnormalities of the spontaneous SPN firing in the alert parkinsonian state. Furthermore, these studies also show altered SPN responses to DA replacement in the advanced parkinsonian state. Clearly, there is yet much to learn about the striatal discharges in PD, but studies using primate models are contributing unique information to advance our understanding of pathophysiologic mechanisms.
Collapse
|
48
|
Jalewa J, Sharma MK, Gengler S, Hölscher C. A novel GLP-1/GIP dual receptor agonist protects from 6-OHDA lesion in a rat model of Parkinson's disease. Neuropharmacology 2017; 117:238-248. [DOI: 10.1016/j.neuropharm.2017.02.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 12/25/2022]
|
49
|
Salvatori I, Valle C, Ferri A, Carrì MT. SIRT3 and mitochondrial metabolism in neurodegenerative diseases. Neurochem Int 2017; 109:184-192. [PMID: 28449871 DOI: 10.1016/j.neuint.2017.04.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/12/2017] [Accepted: 04/21/2017] [Indexed: 02/07/2023]
Abstract
The NAD+-dependent deacetylase protein Sirtuin 3 (SIRT3) is emerging among the factors playing a key role in the regulation of mitochondrial function and in the prevention of oxidative stress. This deacetylase activates protein substrates directly involved in the production and detoxification of ROS, such as superoxide dismutase 2 and catalase, but also enzymes in the lipid beta-oxidation pathway. In this paper we review existing evidence on the role of SIRT3 in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington disease, including data from new experiments in a model for amyotrophic lateral sclerosis linked to mutations in superoxide dismutase 1. Specifically, we report that expression of the mitochondrial isoform of SIRT3 is altered in muscle from the G93A-SOD1 mice during progression of disease; this alteration influences mitochondrial metabolism, which may be relevant for the well known energetic alterations taking place in ALS patients. These data reinforce the concept that SIRT3 may be a relevant therapeutic target is ALS as well as in other neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Cristiana Valle
- Fondazione Santa Lucia IRCCS, Rome, Italy; Institute for Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Alberto Ferri
- Fondazione Santa Lucia IRCCS, Rome, Italy; Institute for Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Maria Teresa Carrì
- Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
50
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|