1
|
Wichmann T, Nelson A, Torres ERS, Svenningsson P, Marongiu R. Leveraging animal models to understand non-motor symptoms of Parkinson's disease. Neurobiol Dis 2025; 208:106848. [PMID: 40023327 DOI: 10.1016/j.nbd.2025.106848] [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: 10/10/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 03/04/2025] Open
Abstract
Parkinson's disease is diagnosed based on motor symptoms, but non-motor symptoms of the disease, such as cognitive impairment, autonomic dysfunction, hyposmia, sleep disorders, and psychiatric disorders heavily impact patient and caregiver quality of life. It has proven challenging to faithfully reproduce and quantify these non-motor phenotypes. Indeed, many non-motor signs in animals that may phenotypically resemble features in patients may be caused by different mechanisms or may not be consistent within the same or similar models. In this review, we survey the existing literature on the assessment of non-motor signs in parkinsonian rodents and non-human primates. We highlight the gaps in our understanding and suggest how researchers might improve experimental designs to produce more meaningful results with the hope of better understanding the disease and developing better therapies.
Collapse
Affiliation(s)
- Thomas Wichmann
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA 30329, USA; Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA 30329, USA; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Alexandra Nelson
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Eileen Ruth S Torres
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Per Svenningsson
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Roberta Marongiu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA; Department of Genetic Medicine, New-York Hospital-Cornell Medical College, New York, NY, USA; Feil Family Brain and Mind Institute, New-York Hospital-Cornell Medical College, New York, NY, USA.
| |
Collapse
|
2
|
Hendrix CM, Baker HE, Yu Y, Schneck DD, Wang J, Johnson LA, Vitek JL. Parkinsonism Disrupts Neuronal Modulation in the Presupplementary Motor Area during Movement Preparation. J Neurosci 2025; 45:e1802242025. [PMID: 39890463 PMCID: PMC11949476 DOI: 10.1523/jneurosci.1802-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/19/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Multiple studies suggest that Parkinson's disease (PD) is associated with changes in neuronal activity throughout the basal ganglia-thalamocortical motor circuit. There are limited electrophysiological data, however, describing how parkinsonism impacts neuronal activity in the presupplementary motor area (pre-SMA), an area in the medial frontal cortex involved in movement planning and motor control. In this study, single unit activity was recorded in the pre-SMA of two female nonhuman primates during a visually cued reaching task in both the naive and parkinsonian state using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of parkinsonism. In the naive state, neuronal discharge rates were dynamically modulated prior to the presentation of the instructional go-cue. In a subset of these modulated cells, the magnitude of modulation correlated linearly with reaction time (RT). In the parkinsonian state, however, modulation of discharge rates in the pre-SMA was disrupted, and the predictive encoding of RT was significantly diminished. These findings add to our understanding of the role of pre-SMA in motor behavior and suggest that disrupted encoding in this cortical region contributes to the alteration of early preparatory and premovement processes present in Parkinson's disease.
Collapse
Affiliation(s)
- Claudia M Hendrix
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Hannah E Baker
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Ying Yu
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - David D Schneck
- University of Minnesota Masonic Institute for the Developing Brain, Minneapolis, Minnesota 55414
| | - Jing Wang
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Luke A Johnson
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Jerrold L Vitek
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
3
|
Poniatowski ŁA, Joniec-Maciejak I, Wawer A, Sznejder-Pachołek A, Machaj E, Ziętal K, Mirowska-Guzel D. Dose-Ranging Effects of the Intracerebral Administration of Atsttrin in Experimental Model of Parkinson's Disease Induced by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in Mice. Mol Neurobiol 2024; 61:9432-9458. [PMID: 38642286 PMCID: PMC11496375 DOI: 10.1007/s12035-024-04161-0] [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: 02/03/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease is one of the most common neurodegenerative disorders characterized by a multitude of motor and non-motor clinical symptoms resulting from the progressive and long-lasting abnormal loss of nigrostriatal dopaminergic neurons. Currently, the available treatments for patients with Parkinson's disease are limited and exert only symptomatic effects, without adequate signs of delaying or stopping the progression of the disease. Atsttrin constitutes the bioengineered protein which ultrastructure is based on the polypeptide chain frame of the progranulin (PGRN), which exerts anti-inflammatory effects through the inhibition of TNFα. The conducted preclinical studies suggest that the therapeutic implementation of Atsttrin may be potentially effective in the treatment of neurodegenerative diseases that are associated with the occurrence of neuroinflammatory processes. The aim of the proposed study was to investigate the effect of direct bilateral intracerebral administration of Atsttrin using stereotactic methods in the preclinical C57BL/6 mouse model of Parkinson's disease inducted by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication. The analysis of the dose dependency effects of the increasing doses of Atsttrin has covered a number of parameters and markers regarding neurodegenerative processes and inflammatory responses including IL-1α, TNFα, IL-6, TH, and TG2 mRNA expressions. Accordingly, the evaluation of the changes in the neurochemical profile included DA, DOPAC, 3-MT, HVA, NA, MHPG, 5-HT, and 5-HIAA concentration levels. The intracerebral administration of Atsttrin into the striatum effectively attenuated the neuroinflammatory reaction in evaluated neuroanatomical structures. Furthermore, the partial restoration of monoamine content and its metabolic turnover were observed. In this case, taking into account the previously described pharmacokinetic profile and extrapolated bioavailability as well as the stability characteristics of Atsttrin, an attempt was made to describe as precisely as possible the quantitative and qualitative effects of increasing doses of the compound within the brain tissue microenvironment in the presented preclinical model of the disease. Collectively, this findings demonstrated that the intracerebral administration of Atsttrin may represent a potential novel therapeutic method for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Łukasz A Poniatowski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Adriana Wawer
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Anna Sznejder-Pachołek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Ewa Machaj
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Katarzyna Ziętal
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
4
|
Chu HY, Smith Y, Lytton WW, Grafton S, Villalba R, Masilamoni G, Wichmann T. Dysfunction of motor cortices in Parkinson's disease. Cereb Cortex 2024; 34:bhae294. [PMID: 39066504 PMCID: PMC11281850 DOI: 10.1093/cercor/bhae294] [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: 02/18/2024] [Revised: 06/26/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The cerebral cortex has long been thought to be involved in the pathophysiology of motor symptoms of Parkinson's disease. The impaired cortical function is believed to be a direct and immediate effect of pathologically patterned basal ganglia output, mediated to the cerebral cortex by way of the ventral motor thalamus. However, recent studies in humans with Parkinson's disease and in animal models of the disease have provided strong evidence suggesting that the involvement of the cerebral cortex is much broader than merely serving as a passive conduit for subcortical disturbances. In the present review, we discuss Parkinson's disease-related changes in frontal cortical motor regions, focusing on neuropathology, plasticity, changes in neurotransmission, and altered network interactions. We will also examine recent studies exploring the cortical circuits as potential targets for neuromodulation to treat Parkinson's disease.
Collapse
Affiliation(s)
- Hong-Yuan Chu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Rd N.W., Washington D.C. 20007, United States
| | - Yoland Smith
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Department of Neurology, School of Medicine, Emory University, 12 Executive Drive N.E., Atlanta, GA 30329, United States
- Emory National Primate Research Center, 954 Gatewood Road N.E., Emory University, Atlanta, GA 30329, United States
| | - William W Lytton
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Department of Physiology & Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, United States
- Department of Neurology, Kings County Hospital, 451 Clarkson Avenue,Brooklyn, NY 11203, United States
| | - Scott Grafton
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Department of Psychological and Brain Sciences, University of California, 551 UCEN Road, Santa Barbara, CA 93106, United States
| | - Rosa Villalba
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Emory National Primate Research Center, 954 Gatewood Road N.E., Emory University, Atlanta, GA 30329, United States
| | - Gunasingh Masilamoni
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Emory National Primate Research Center, 954 Gatewood Road N.E., Emory University, Atlanta, GA 30329, United States
| | - Thomas Wichmann
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States
- Department of Neurology, School of Medicine, Emory University, 12 Executive Drive N.E., Atlanta, GA 30329, United States
- Emory National Primate Research Center, 954 Gatewood Road N.E., Emory University, Atlanta, GA 30329, United States
| |
Collapse
|
5
|
Boi L, Johansson Y, Tonini R, Moratalla R, Fisone G, Silberberg G. Serotonergic and dopaminergic neurons in the dorsal raphe are differentially altered in a mouse model for parkinsonism. eLife 2024; 12:RP90278. [PMID: 38940422 PMCID: PMC11213571 DOI: 10.7554/elife.90278] [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] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is characterized by motor impairments caused by degeneration of dopamine neurons in the substantia nigra pars compacta. In addition to these symptoms, PD patients often suffer from non-motor comorbidities including sleep and psychiatric disturbances, which are thought to depend on concomitant alterations of serotonergic and noradrenergic transmission. A primary locus of serotonergic neurons is the dorsal raphe nucleus (DRN), providing brain-wide serotonergic input. Here, we identified electrophysiological and morphological parameters to classify serotonergic and dopaminergic neurons in the murine DRN under control conditions and in a PD model, following striatal injection of the catecholamine toxin, 6-hydroxydopamine (6-OHDA). Electrical and morphological properties of both neuronal populations were altered by 6-OHDA. In serotonergic neurons, most changes were reversed when 6-OHDA was injected in combination with desipramine, a noradrenaline (NA) reuptake inhibitor, protecting the noradrenergic terminals. Our results show that the depletion of both NA and dopamine in the 6-OHDA mouse model causes changes in the DRN neural circuitry.
Collapse
Affiliation(s)
- Laura Boi
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | - Yvonne Johansson
- Department of Neuroscience, Karolinska InstituteStockholmSweden
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College LondonLondonUnited Kingdom
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di TecnologiaGenovaItaly
| | - Rosario Moratalla
- Cajal Institute, Spanish National Research Council (CSIC)MadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | | |
Collapse
|
6
|
Norris SA, Tian L, Williams EL, Perlmutter JS. Transient dystonia correlates with parkinsonism after 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine in nonhuman primates. DYSTONIA (LAUSANNE, SWITZERLAND) 2023; 2:11019. [PMID: 37711667 PMCID: PMC10501383 DOI: 10.3389/dyst.2023.11019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Unilateral internal carotid artery 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) infusion in non-human primates produces transient contralateral hemi-dystonia followed by stable contralateral hemi-parkinsonism; the relationship between dystonia and parkinsonism remains unclear. We hypothesized that transient dystonia severity following MPTP correlates with parkinsonism severity. In male Macaca nemestrina (n = 3) and M. fascicularis (n = 17) we administered unilateral intra-carotid MPTP, then correlated validated blinded ratings of transient peak dystonia and delayed parkinsonism. We also correlated dystonia severity with post-mortem measures of residual striatal dopamine and nigral neuron counts obtained a mean 53 ± 15 days following MPTP, after resolution of dystonia but during stable parkinsonism. Median latency to dystonia onset was 1 day, and peak severity 2.5 days after MPTP; total dystonia duration was 13.5 days. Parkinsonism peaked a median of 19.5 days after MPTP, remaining nearly constant thereafter. Peak dystonia severity highly correlated with parkinsonism severity (r[18] = 0.82, p < 0.001). Residual cell counts in lesioned nigra correlated linearly with peak dystonia scores (r[18] = -0.68, p=<0.001). Dystonia was not observed in monkeys without striatal dopamine depletion (n = 2); dystonia severity correlated with striatal dopamine depletion when residual nigral cell loss was less than 50% ([11] r = -0.83, p < 0.001) but spanned a broad range with near complete striatal dopamine depletion, when nigral cell loss was greater than 50%. Our data indicate that residual striatal dopamine may not reflect dystonia severity. We speculate on mechanisms of transient dystonia followed by parkinsonism that may be studied using this particular NHP MPTP model to better understand relationships of transient dystonia to nigrostriatal injury and parkinsonism.
Collapse
Affiliation(s)
- S. A. Norris
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - L. Tian
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - E. L. Williams
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - J. S. Perlmutter
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neuroscience, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Physical Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Occupational Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
7
|
Dorninger F, Kiss A, Rothauer P, Stiglbauer-Tscholakoff A, Kummer S, Fallatah W, Perera-Gonzalez M, Hamza O, König T, Bober MB, Cavallé-Garrido T, Braverman NE, Forss-Petter S, Pifl C, Bauer J, Bittner RE, Helbich TH, Podesser BK, Todt H, Berger J. Overlapping and Distinct Features of Cardiac Pathology in Inherited Human and Murine Ether Lipid Deficiency. Int J Mol Sci 2023; 24:1884. [PMID: 36768204 PMCID: PMC9914995 DOI: 10.3390/ijms24031884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Inherited deficiency in ether lipids, a subgroup of glycerophospholipids with unique biochemical and biophysical properties, evokes severe symptoms in humans resulting in a multi-organ syndrome. Mouse models with defects in ether lipid biosynthesis have widely been used to understand the pathophysiology of human disease and to study the roles of ether lipids in various cell types and tissues. However, little is known about the function of these lipids in cardiac tissue. Previous studies included case reports of cardiac defects in ether-lipid-deficient patients, but a systematic analysis of the impact of ether lipid deficiency on the mammalian heart is still missing. Here, we utilize a mouse model of complete ether lipid deficiency (Gnpat KO) to accomplish this task. Similar to a subgroup of human patients with rhizomelic chondrodysplasia punctata (RCDP), a fraction of Gnpat KO fetuses present with defects in ventricular septation, presumably evoked by a developmental delay. We did not detect any signs of cardiomyopathy but identified increased left ventricular end-systolic and end-diastolic pressure in middle-aged ether-lipid-deficient mice. By comprehensive electrocardiographic characterization, we consistently found reduced ventricular conduction velocity, as indicated by a prolonged QRS complex, as well as increased QRS and QT dispersion in the Gnpat KO group. Furthermore, a shift of the Wenckebach point to longer cycle lengths indicated depressed atrioventricular nodal function. To complement our findings in mice, we analyzed medical records and performed electrocardiography in ether-lipid-deficient human patients, which, in contrast to the murine phenotype, indicated a trend towards shortened QT intervals. Taken together, our findings demonstrate that the cardiac phenotype upon ether lipid deficiency is highly heterogeneous, and although the manifestations in the mouse model only partially match the abnormalities in human patients, the results add to our understanding of the physiological role of ether lipids and emphasize their importance for proper cardiac development and function.
Collapse
Affiliation(s)
- Fabian Dorninger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Attila Kiss
- Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Peter Rothauer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstrasse 13a, 1090 Vienna, Austria
| | - Alexander Stiglbauer-Tscholakoff
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Stefan Kummer
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Währinger Straße 13, 1090 Vienna, Austria
| | - Wedad Fallatah
- Department of Genetic Medicine, King AbdulAziz University, Jeddah 21589, Saudi Arabia
- Department of Human Genetics and Pediatrics, Montreal Children’s Hospital, McGill University, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Mireia Perera-Gonzalez
- Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Ouafa Hamza
- Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Theresa König
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Michael B. Bober
- Skeletal Dysplasia Program, Nemours Children’s Hospital, 1600 Rockland Road, Wilmington, DE 19803, USA
| | - Tiscar Cavallé-Garrido
- Department of Pediatrics, Division of Cardiology, Montreal Children’s Hospital, McGill University, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Nancy E. Braverman
- Department of Human Genetics and Pediatrics, Montreal Children’s Hospital, McGill University, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Christian Pifl
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Reginald E. Bittner
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Währinger Straße 13, 1090 Vienna, Austria
| | - Thomas H. Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Bruno K. Podesser
- Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Hannes Todt
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstrasse 13a, 1090 Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| |
Collapse
|
8
|
Parr-Brownlie LC, Itoga CA, Walters JR, Underwood CF. Oscillatory waveform sharpness asymmetry changes in motor thalamus and motor cortex in a rat model of Parkinson's disease. Exp Neurol 2022; 354:114089. [PMID: 35461830 PMCID: PMC11345867 DOI: 10.1016/j.expneurol.2022.114089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/17/2022] [Indexed: 11/23/2022]
Abstract
Parkinson's disease (PD) causes bursty and oscillatory activity in basal ganglia output that is thought to contribute to movement deficits through impact on motor thalamus and motor cortex (MCx). We examined the effect of dopamine loss on motor thalamus and motor cortex activity by recording neuronal and LFP activities in ventroanterior-ventrolateral (VAVL) thalamus and MCx in urethane-anesthetised control and parkinsonian rats. Dopamine lesion decreased the firing rate and increased the bursting of putative pyramidal neurons in layer V, but not layer VI, of the MCx without changing other aspects of firing pattern. In contrast, dopamine lesion did not affect VAVL firing rate, pattern or low threshold calcium spike bursts. Slow-wave (~1 Hz) oscillations in LFP recordings were analyzed with conventional power and waveform shape analyses. While dopamine lesion did not influence total power, it was consistently associated with an increase in oscillatory waveform sharpness asymmetry (i.e., sharper troughs vs. peaks) in both motor thalamus and MCx. Furthermore, we found that measures of sharpness asymmetry were positively correlated in paired motor thalamus-MCx recordings, and that correlation coefficients were larger in dopamine lesioned rats. These data support the idea that dysfunctional MCx activity in parkinsonism emerges from subsets of cell groups (e.g. layer V pyramidal neurons) and is evident in the shape but not absolute power of slow-wave oscillations. Hypoactive layer V pyramidal neuron firing in dopamine lesioned rats is unlikely to be driven by VAVL thalamus and may, therefore, reflect the loss of mesocortical dopaminergic afferents and/or changes in intrinsic excitability.
Collapse
Affiliation(s)
- Louise C Parr-Brownlie
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand; Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Building 35 Room 1C 903, Bethesda, MD 20892-3702, USA.
| | - Christy A Itoga
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Building 35 Room 1C 903, Bethesda, MD 20892-3702, USA
| | - Judith R Walters
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Building 35 Room 1C 903, Bethesda, MD 20892-3702, USA
| | - Conor F Underwood
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| |
Collapse
|
9
|
Norris SA, White H, Tanenbaum A, Williams EL, Cruchaga C, Tian L, Schmidt RE, Perlmutter JS. Severe acute neurotoxicity reflects absolute intra-carotid 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine dose in non-human primates. J Neurosci Methods 2022; 366:109406. [PMID: 34767855 DOI: 10.1016/j.jneumeth.2021.109406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 11/18/2022]
Affiliation(s)
- S A Norris
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA; Departments of Radiology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA.
| | - Hcb White
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - A Tanenbaum
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - E L Williams
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - C Cruchaga
- Departments of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - L Tian
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - R E Schmidt
- Departments of Pathology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - J S Perlmutter
- Departments of Neurology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA; Departments of Radiology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA; Departments of Neuroscience, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA; Departments of Physical, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA; Departments of Occupational Therapy, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Mizrahi-Kliger AD, Feldmann LK, Kühn AA, Bergman H. Etiologies of insomnia in Parkinson's disease - Lessons from human studies and animal models. Exp Neurol 2022; 350:113976. [PMID: 35026228 DOI: 10.1016/j.expneurol.2022.113976] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/27/2021] [Accepted: 01/06/2022] [Indexed: 12/28/2022]
Abstract
Sleep disorders are integral to Parkinson's disease (PD). Insomnia, an inability to maintain stable sleep, affects most patients and is widely rated as one of the most debilitating facets of this disease. PD insomnia is often perceived as a multifactorial entity - a consequence of several of the disease symptoms, comorbidities and therapeutic strategies. Yet, this view evolved against a backdrop of a relative scarcity of works trying to directly dissect the underlying neural correlates and mechanisms in animal models. The last years have seen the emergence of a wealth of new evidence regarding the neural underpinnings of insomnia in PD. Here, we review early and recent reports from patients and animal models evaluating the etiology of PD insomnia. We start by outlining the phenomenology of PD insomnia and continue to analyze the evidence supporting insomnia as emanating from four distinct subdivisions of etiologies - the symptoms and comorbidities of the disease, the medical therapy, the degeneration of non-dopaminergic cell groups and subsequent alterations in circadian rhythms, and the degeneration of dopaminergic neurons in the brainstem and its resulting effect on the basal ganglia. Finally, we review emerging neuromodulation-based therapeutic avenues for PD insomnia.
Collapse
Affiliation(s)
- Aviv D Mizrahi-Kliger
- Department of Neurobiology, Institute of Medical Research Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| | - Lucia K Feldmann
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin 10117, Germany
| | - Andrea A Kühn
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin 10117, Germany; NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin, Germany
| | - Hagai Bergman
- Department of Neurobiology, Institute of Medical Research Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Neurosurgery, Hadassah University Hospital, Jerusalem 91120, Israel
| |
Collapse
|
11
|
Lama J, Buhidma Y, Fletcher E, Duty S. Animal models of Parkinson's disease: a guide to selecting the optimal model for your research. Neuronal Signal 2021; 5:NS20210026. [PMID: 34956652 PMCID: PMC8661507 DOI: 10.1042/ns20210026] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a complex, multisystem disorder characterised by α-synuclein (SNCA) pathology, degeneration of nigrostriatal dopaminergic neurons, multifactorial pathogenetic mechanisms and expression of a plethora of motor and non-motor symptoms. Animal models of PD have already been instructive in helping us unravel some of these aspects. However, much remains to be discovered, requiring continued interrogation by the research community. In contrast with the situation for many neurological disorders, PD benefits from of a wide range of available animal models (pharmacological, toxin, genetic and α-synuclein) but this makes selection of the optimal one for a given study difficult. This is especially so when a study demands a model that displays a specific combination of features. While many excellent reviews of animal models already exist, this review takes a different approach with the intention of more readily informing this decision-making process. We have considered each feature of PD in turn - aetiology, pathology, pathogenesis, motor dysfunctions and non-motor symptoms (NMS) - highlighting those animal models that replicate each. By compiling easily accessible tables and a summary figure, we aim to provide the reader with a simple, go-to resource for selecting the optimal animal model of PD to suit their research needs.
Collapse
Affiliation(s)
- Joana Lama
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Yazead Buhidma
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Edward J.R. Fletcher
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| | - Susan Duty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age Related Diseases, Wolfson Wing, Hodgkin Building, Guy’s Campus, London SE1 1UL, U.K
| |
Collapse
|
12
|
Masilamoni GJ, Weinkle A, Papa SM, Smith Y. Cortical Serotonergic and Catecholaminergic Denervation in MPTP-Treated Parkinsonian Monkeys. Cereb Cortex 2021; 32:1804-1822. [PMID: 34519330 DOI: 10.1093/cercor/bhab313] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 11/14/2022] Open
Abstract
Decreased cortical serotonergic and catecholaminergic innervation of the frontal cortex has been reported at early stages of Parkinson's disease (PD). However, the limited availability of animal models that exhibit these pathological features has hampered our understanding of the functional significance of these changes during the course of the disease. In the present study, we assessed longitudinal changes in cortical serotonin and catecholamine innervation in motor-symptomatic and asymptomatic monkeys chronically treated with low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Densitometry and unbiased stereological techniques were used to quantify changes in serotonin and tyrosine hydroxylase (TH) immunoreactivity in frontal cortices of 3 control monkeys and 3 groups of MPTP-treated monkeys (motor-asymptomatic [N = 2], mild parkinsonian [N = 3], and moderate parkinsonian [N = 3]). Our findings revealed a significant decrease (P < 0.001) in serotonin innervation of motor (Areas 4 and 6), dorsolateral prefrontal (Areas 9 and 46), and limbic (Areas 24 and 25) cortical areas in motor-asymptomatic MPTP-treated monkeys. Both groups of symptomatic MPTP-treated animals displayed further serotonin denervation in these cortical regions (P < 0.0001). A significant loss of serotonin-positive dorsal raphe neurons was found in the moderate parkinsonian group. On the other hand, the intensity of cortical TH immunostaining was not significantly affected in motor asymptomatic MPTP-treated monkeys, but underwent a significant reduction in the moderate symptomatic group (P < 0.05). Our results indicate that chronic intoxication with MPTP induces early pathology in the corticopetal serotonergic system, which may contribute to early non-motor symptoms in PD.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj Masilamoni
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allison Weinkle
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Neuroplasticity and Multilevel System of Connections Determine the Integrative Role of Nucleus Accumbens in the Brain Reward System. Int J Mol Sci 2021; 22:ijms22189806. [PMID: 34575969 PMCID: PMC8471564 DOI: 10.3390/ijms22189806] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
A growing body of evidence suggests that nucleus accumbens (NAc) plays a significant role not only in the physiological processes associated with reward and satisfaction but also in many diseases of the central nervous system. Summary of the current state of knowledge on the morphological and functional basis of such a diverse function of this structure may be a good starting point for further basic and clinical research. The NAc is a part of the brain reward system (BRS) characterized by multilevel organization, extensive connections, and several neurotransmitter systems. The unique role of NAc in the BRS is a result of: (1) hierarchical connections with the other brain areas, (2) a well-developed morphological and functional plasticity regulating short- and long-term synaptic potentiation and signalling pathways, (3) cooperation among several neurotransmitter systems, and (4) a supportive role of neuroglia involved in both physiological and pathological processes. Understanding the complex function of NAc is possible by combining the results of morphological studies with molecular, genetic, and behavioral data. In this review, we present the current views on the NAc function in physiological conditions, emphasizing the role of its connections, neuroplasticity processes, and neurotransmitter systems.
Collapse
|
14
|
Bhat S, Niello M, Schicker K, Pifl C, Sitte HH, Freissmuth M, Sandtner W. Handling of intracellular K + determines voltage dependence of plasmalemmal monoamine transporter function. eLife 2021; 10:67996. [PMID: 34061030 PMCID: PMC8192120 DOI: 10.7554/elife.67996] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/30/2021] [Indexed: 12/16/2022] Open
Abstract
The concentrative power of the transporters for dopamine (DAT), norepinephrine (NET), and serotonin (SERT) is thought to be fueled by the transmembrane Na+ gradient, but it is conceivable that they can also tap other energy sources, for example, membrane voltage and/or the transmembrane K+ gradient. We have addressed this by recording uptake of endogenous substrates or the fluorescent substrate APP+(4-(4-dimethylamino)phenyl-1-methylpyridinium) under voltage control in cells expressing DAT, NET, or SERT. We have shown that DAT and NET differ from SERT in intracellular handling of K+. In DAT and NET, substrate uptake was voltage-dependent due to the transient nature of intracellular K+ binding, which precluded K+ antiport. SERT, however, antiports K+ and achieves voltage-independent transport. Thus, there is a trade-off between maintaining constant uptake and harvesting membrane potential for concentrative power, which we conclude to occur due to subtle differences in the kinetics of co-substrate ion binding in closely related transporters.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus Schicker
- Division of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
El-Gamal M, Salama M, Collins-Praino LE, Baetu I, Fathalla AM, Soliman AM, Mohamed W, Moustafa AA. Neurotoxin-Induced Rodent Models of Parkinson's Disease: Benefits and Drawbacks. Neurotox Res 2021; 39:897-923. [PMID: 33765237 DOI: 10.1007/s12640-021-00356-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by cardinal motor impairments, including akinesia and tremor, as well as by a host of non-motor symptoms, including both autonomic and cognitive dysfunction. PD is associated with a death of nigral dopaminergic neurons, as well as the pathological spread of Lewy bodies, consisting predominantly of the misfolded protein alpha-synuclein. To date, only symptomatic treatments, such as levodopa, are available, and trials aiming to cure the disease, or at least halt its progression, have not been successful. Wong et al. (2019) suggested that the lack of effective therapy against neurodegeneration in PD might be attributed to the fact that the molecular mechanisms standing behind the dopaminergic neuronal vulnerability are still a major scientific challenge. Understanding these molecular mechanisms is critical for developing effective therapy. Thirty-five years ago, Calne and William Langston (1983) raised the question of whether biological or environmental factors precipitate the development of PD. In spite of great advances in technology and medicine, this question still lacks a clear answer. Only 5-15% of PD cases are attributed to a genetic mutation, with the majority of cases classified as idiopathic, which could be linked to exposure to environmental contaminants. Rodent models play a crucial role in understanding the risk factors and pathogenesis of PD. Additionally, well-validated rodent models are critical for driving the preclinical development of clinically translatable treatment options. In this review, we discuss the mechanisms, similarities and differences, as well as advantages and limitations of different neurotoxin-induced rat models of PD. In the second part of this review, we will discuss the potential future of neurotoxin-induced models of PD. Finally, we will briefly demonstrate the crucial role of gene-environment interactions in PD and discuss fusion or dual PD models. We argue that these models have the potential to significantly further our understanding of PD.
Collapse
Affiliation(s)
- Mohamed El-Gamal
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohamed Salama
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), Dublin, Ireland
| | | | | | - Ahmed M Fathalla
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amira M Soliman
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wael Mohamed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Mansoura, Egypt.,Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University, Kuantan, Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and Marcs Institute for Brain and Behaviour, Western Sydney University, Sydney, NSW, Australia.,Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
16
|
Globus pallidus, but not entopeduncular nucleus, 6-OHDA-induced lesion attenuates L-Dopa-induced dyskinesia in the rat model of Parkinson's disease. Pharmacol Biochem Behav 2020; 197:173013. [DOI: 10.1016/j.pbb.2020.173013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
|
17
|
Sgambato V. Breathing new life into neurotoxic-based monkey models of Parkinson's disease to study the complex biological interplay between serotonin and dopamine. PROGRESS IN BRAIN RESEARCH 2020; 261:265-285. [PMID: 33785131 DOI: 10.1016/bs.pbr.2020.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous clinical studies have shown that the serotonergic system also degenerates in patients with Parkinson's disease. The causal role of this impairment in Parkinson's symptomatology and the response to treatment remains to be refined, in particular thanks to approaches allowing the two components DA and 5-HT to be isolated if possible. We have developed a macaque monkey model of Parkinson's disease exhibiting a double lesion (dopaminergic and serotonergic) thanks to the sequential use of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and MDMA (3,4-methylenedioxy-N-methamphetamine) (or MDMA prior MPTP). We characterized this monkey model by multimodal imaging (PET, positron emission tomography with several radiotracers; DTI, diffusion tensor imaging), behavioral assessments (parkinsonism, dyskinesia, neuropsychiatric-like behavior) and post-mortem analysis (with DA and 5-HT markers). When administrated after MPTP, MDMA damaged the 5-HT presynaptic system without affecting the remaining DA neurons. The lesion of 5-HT fibers induced by MDMA altered rigidity and prevented dyskinesia and neuropsychiatric-like symptoms induced by levodopa therapy in MPTP-treated animals. Interestingly also, prior MDMA administration aggravates the parkinsonian deficits and associated DA injury. Dystonic postures, action tremor and global spontaneous activities were significantly affected. All together, these data clearly indicate that late or early lesions of the 5-HT system have a differential impact on parkinsonian symptoms in the macaque model of Parkinson's disease. Whether MDMA has an impact on neuropsychiatric-like symptoms such as apathy, anxiety, depression remains to be addressed. Despite its limitations, this toxin-based double-lesioned monkey model takes on its full meaning and provides material for the experimental study of the heterogeneity of patients.
Collapse
Affiliation(s)
- Véronique Sgambato
- Université de Lyon, CNRS UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.
| |
Collapse
|
18
|
Bharatiya R, Chagraoui A, De Deurwaerdere S, Argiolas A, Melis MR, Sanna F, De Deurwaerdere P. Chronic Administration of Fipronil Heterogeneously Alters the Neurochemistry of Monoaminergic Systems in the Rat Brain. Int J Mol Sci 2020; 21:ijms21165711. [PMID: 32784929 PMCID: PMC7461054 DOI: 10.3390/ijms21165711] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
Fipronil (FPN), a widely used pesticide for agricultural and non-agricultural pest control, is possibly neurotoxic for mammals. Brain monoaminergic systems, involved in virtually all brain functions, have been shown to be sensitive to numerous pesticides. Here, we addressed the hypothesis that chronic exposure to FPN could modify brain monoamine neurochemistry. FPN (10 mg/kg) was chronically administered for 21 days through oral gavage in rats. Thereafter, the tissue concentrations of dopamine (DA) and its metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid; serotonin (5-HT) and its metabolite, 5-hydroxyindoleacetic acid (5-HIAA); and noradrenaline (NA) were measured in 30 distinct brain regions. FPN significantly decreased DA and its metabolite levels in most striatal territories, including the nucleus accumbens and the substantia nigra (SN). FPN also diminished 5-HT levels in some striatal regions and the SN. The indirect index of the turnovers, DOPAC/DA and 5-HIAA/5-HT ratios, was increased in numerous brain regions. FPN reduced the NA content only in the nucleus accumbens core. Using the Bravais–Pearson test to study the neurochemical organization of monoamines through multiple correlative analyses across the brain, we found fewer correlations for NA, DOPAC/DA, and 5-HIAA/5-HT ratios, and an altered pattern of correlations within and between monoamine systems. We therefore conclude that the chronic administration of FPN in rats induces massive and inhomogeneous changes in the DA and 5-HT systems in the brain.
Collapse
Affiliation(s)
- Rahul Bharatiya
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09100 Cagliari, Italy; (R.B.); (A.A.); (M.R.M.)
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 146 rue Léo Saignat, B.P.281, F-33000 Bordeaux CEDEX, France;
| | - Abdeslam Chagraoui
- Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), Normandie Univ, UNIROUEN, INSERM, U1239, CHU Rouen, 76000 Rouen, France;
- Department of Medical Biochemistry, Rouen University Hospital, 76000 Rouen, France
| | - Salomé De Deurwaerdere
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 146 rue Léo Saignat, B.P.281, F-33000 Bordeaux CEDEX, France;
| | - Antonio Argiolas
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09100 Cagliari, Italy; (R.B.); (A.A.); (M.R.M.)
- Centre of Excellence for the Neurobiology of Addictions, University of Cagliari, 09100 Cagliari, Italy
- Institute of Neuroscience, National Research Council, Cagliari Section, University of Cagliari, 09100 Cagliari, Italy
| | - Maria Rosaria Melis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09100 Cagliari, Italy; (R.B.); (A.A.); (M.R.M.)
- Centre of Excellence for the Neurobiology of Addictions, University of Cagliari, 09100 Cagliari, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09100 Cagliari, Italy; (R.B.); (A.A.); (M.R.M.)
- Correspondence: (F.S.); (P.D.D.); Tel.: +39-070-675-4330 (F.S.); +33-557-571-290 (P.D.D.)
| | - Philippe De Deurwaerdere
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5287), 146 rue Léo Saignat, B.P.281, F-33000 Bordeaux CEDEX, France;
- Correspondence: (F.S.); (P.D.D.); Tel.: +39-070-675-4330 (F.S.); +33-557-571-290 (P.D.D.)
| |
Collapse
|
19
|
|
20
|
Basal ganglia beta oscillations during sleep underlie Parkinsonian insomnia. Proc Natl Acad Sci U S A 2020; 117:17359-17368. [PMID: 32636265 DOI: 10.1073/pnas.2001560117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sleep disorders are among the most debilitating comorbidities of Parkinson's disease (PD) and affect the majority of patients. Of these, the most common is insomnia, the difficulty to initiate and maintain sleep. The degree of insomnia correlates with PD severity and it responds to treatments that decrease pathological basal ganglia (BG) beta oscillations (10-17 Hz in primates), suggesting that beta activity in the BG may contribute to insomnia. We used multiple electrodes to record BG spiking and field potentials during normal sleep and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism in nonhuman primates. MPTP intoxication resulted in severe insomnia with delayed sleep onset, sleep fragmentation, and increased wakefulness. Insomnia was accompanied by the onset of nonrapid eye movement (NREM) sleep beta oscillations that were synchronized across the BG and cerebral cortex. The BG beta oscillatory activity was associated with a decrease in slow oscillations (0.1-2 Hz) throughout the cortex, and spontaneous awakenings were preceded by an increase in BG beta activity and cortico-BG beta coherence. Finally, the increase in beta oscillations in the basal ganglia during sleep paralleled decreased NREM sleep, increased wakefulness, and more frequent awakenings. These results identify NREM sleep beta oscillation in the BG as a neural correlate of PD insomnia and suggest a mechanism by which this disorder could emerge.
Collapse
|
21
|
Paredes-Rodriguez E, Vegas-Suarez S, Morera-Herreras T, De Deurwaerdere P, Miguelez C. The Noradrenergic System in Parkinson's Disease. Front Pharmacol 2020; 11:435. [PMID: 32322208 PMCID: PMC7157437 DOI: 10.3389/fphar.2020.00435] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Nowadays it is well accepted that in Parkinson’s disease (PD), the neurodegenerative process occurs in stages and that damage to other areas precedes the neuronal loss in the substantia nigra pars compacta, which is considered a pathophysiological hallmark of PD. This heterogeneous and progressive neurodegeneration may explain the diverse symptomatology of the disease, including motor and non-motor alterations. In PD, one of the first areas undergoing degeneration is the locus coeruleus (LC). This noradrenergic nucleus provides extensive innervation throughout the brain and plays a fundamental neuromodulator role, participating in stress responses, emotional memory, and control of motor, sensory, and autonomic functions. Early in the disease, LC neurons suffer modifications that can condition the effectiveness of pharmacological treatments, and importantly, can lead to the appearance of common non-motor symptomatology. The noradrenergic system also exerts anti-inflammatory and neuroprotective effect on the dopaminergic degeneration and noradrenergic damage can consequently condition the progress of the disease. From the pharmacological point of view, it is also important to understand how the noradrenergic system performs in PD, since noradrenergic medication is often used in these patients, and drug interactions can take place when combining them with the gold standard drug therapy in PD, L-3,4-dihydroxyphenylalanine (L-DOPA). This review provides an overview about the functional status of the noradrenergic system in PD and its contribution to the efficacy of pharmacological-based treatments. Based on preclinical and clinical publications, a special attention will be dedicated to the most prevalent non-motor symptoms of the disease.
Collapse
Affiliation(s)
- Elena Paredes-Rodriguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Sergio Vegas-Suarez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Philippe De Deurwaerdere
- Centre National de la Recherche scientifique, Institut des Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA UMR 5287), Bordeaux, France
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
22
|
Bacqué-Cazenave J, Bharatiya R, Barrière G, Delbecque JP, Bouguiyoud N, Di Giovanni G, Cattaert D, De Deurwaerdère P. Serotonin in Animal Cognition and Behavior. Int J Mol Sci 2020; 21:ijms21051649. [PMID: 32121267 PMCID: PMC7084567 DOI: 10.3390/ijms21051649] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/20/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is acknowledged as a major neuromodulator of nervous systems in both invertebrates and vertebrates. It has been proposed for several decades that it impacts animal cognition and behavior. In spite of a completely distinct organization of the 5-HT systems across the animal kingdom, several lines of evidence suggest that the influences of 5-HT on behavior and cognition are evolutionary conserved. In this review, we have selected some behaviors classically evoked when addressing the roles of 5-HT on nervous system functions. In particular, we focus on the motor activity, arousal, sleep and circadian rhythm, feeding, social interactions and aggressiveness, anxiety, mood, learning and memory, or impulsive/compulsive dimension and behavioral flexibility. The roles of 5-HT, illustrated in both invertebrates and vertebrates, show that it is more able to potentiate or mitigate the neuronal responses necessary for the fine-tuning of most behaviors, rather than to trigger or halt a specific behavior. 5-HT is, therefore, the prototypical neuromodulator fundamentally involved in the adaptation of all organisms across the animal kingdom.
Collapse
Affiliation(s)
- Julien Bacqué-Cazenave
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
| | - Rahul Bharatiya
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09100 Cagliari, Italy
| | - Grégory Barrière
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
| | - Jean-Paul Delbecque
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
| | - Nouhaila Bouguiyoud
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta;
- School of Biosciences, Neuroscience Division, Cardiff University, Cardiff CF24 4HQ, UK
| | - Daniel Cattaert
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
- Correspondence: (D.C.); (P.D.D.)
| | - Philippe De Deurwaerdère
- INCIA, UMR5287, Centre National de la Recherche Scientifique, 33076 Bordeaux, France; (J.B.-C.); (R.B.); (G.B.); (J.-P.D.); (N.B.)
- Correspondence: (D.C.); (P.D.D.)
| |
Collapse
|
23
|
De Deurwaerdere P, Gaetani S, Vaughan RA. Old neurochemical markers, new functional directions?: An Editorial for 'Distinct gradients of various neurotransmitter markers in caudate nucleus and putamen of the human brain' on page 650. J Neurochem 2020; 152:623-626. [PMID: 31917872 DOI: 10.1111/jnc.14929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
The dorsal striatum coordinates input-output processing of numerous functions including those related to motor activity, motivation, and learning. Considerable anatomical and biochemical heterogeneity across striatal subregions has long been known to result in distinct functional outcomes, and for imbalances in these pathways to contribute to many complex disorders. Here we highlight the study of Hörtnagl et al. (2019) who utilize precision dissection of human caudate nucleus and putamen for detailed measurement of major neurochemical markers to address the question of anatomical heterogeneity of neurotransmitter distribution and turnover in these regions. The findings identify gradients of neurotransmitter markers in rostro-caudal, dorso-lateral, and anterior-posterior directions with a precision that has not been previously determined in humans. Correlative analyses of the results also suggest tentative links between content of various neurotransmitters in specific subregions, raising the intriguing possibility that neurotransmitter quantity in one territory may correlate with the quantity of the same or different transmitter from another territory. This suggests the presence of a functional anatomy over extensive brain regions and networks that can be studied through multiple correlative analyses, and identify a possible basis for a new approach for postmortem analysis of neurotransmitter distribution and function.
Collapse
Affiliation(s)
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
24
|
Monje MHG, Blesa J, García-Cabezas MÁ, Obeso JA, Cavada C. Changes in thalamic dopamine innervation in a progressive Parkinson's disease model in monkeys. Mov Disord 2019; 35:419-430. [PMID: 31800134 PMCID: PMC7154739 DOI: 10.1002/mds.27921] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Dopamine loss beyond the mesostriatal system might be relevant in pathogenic mechanisms and some clinical manifestations in PD. The primate thalamus is densely and heterogeneously innervated with dopaminergic axons, most of which express the dopamine transporter, as does the nigrostriatal system. We hypothesized that dopamine depletion may be present in the thalamus of the parkinsonian brain and set out to ascertain possible regional differences. METHODS The toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine was administered to adult macaque monkeys using a slow intoxication protocol. The treated macaques were classified into 2 groups according to their motor status: nonsymptomatic and parkinsonian. Dopamine innervation was studied with immunohistochemistry for the dopamine transporter. Topographic maps of the dopamine transporter-immunoreactive axon distribution were generated and the total length and length density of these axons stereologically estimated using a 3-dimensional fractionator. RESULTS Parkinsonian macaques exhibited lower dopamine transporter-immunoreactive axon length density than controls in mediodorsal and centromedian-parafascicular nuclei. Dopamine denervation in the mediodorsal nucleus was already noticeable in nonsymptomatic macaques and was even greater in parkinsonian macaques. Reticular nucleus dopamine transporter-immunoreactive axon length density presented an inverse pattern, increasing progressively to the maximum density seen in parkinsonian macaques. No changes were observed in ventral thalamic nuclei. Dopamine transporter-immunoreactive axon maps supported the quantitative findings. CONCLUSIONS Changes in the dopamine innervation of various thalamic nuclei are heterogeneous and start in the premotor parkinsonian stage. These changes may be involved in some poorly understood nonmotor manifestations of PD. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Mariana H G Monje
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,HM-CINAC, HM Puerta del Sur University Hospital, Móstoles, and CEU-San Pablo University, Madrid, Spain
| | - Javier Blesa
- HM-CINAC, HM Puerta del Sur University Hospital, Móstoles, and CEU-San Pablo University, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Miguel Ángel García-Cabezas
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - José A Obeso
- HM-CINAC, HM Puerta del Sur University Hospital, Móstoles, and CEU-San Pablo University, Madrid, Spain.,CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Carmen Cavada
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
25
|
Jiménez-Sánchez L, Blesa J, Del Rey NL, Monje MHG, Obeso JA, Cavada C. Serotonergic innervation of the striatum in a nonhuman primate model of Parkinson's disease. Neuropharmacology 2019; 170:107806. [PMID: 31589886 DOI: 10.1016/j.neuropharm.2019.107806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 10/25/2022]
Abstract
Parkinson's disease (PD) is characterized by dopaminergic neurodegeneration in the substantia nigra and dopamine depletion in the striatum. Non-dopaminergic systems are also affected, including the serotonergic system. Enhanced striatal serotonergic innervation is a proposed compensatory mechanism for the dopaminergic deficit. Meanwhile a serotonergic deficit has been suggested as preceding the nigrostriatal dopaminergic pathology in PD. Our aim was to assess the serotonergic innervation of the striatum in a model of progressive experimental parkinsonism in macaques, from pre-symptomatic to symptomatic stages. The neurotoxin 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) was administered to adult macaque monkeys using a slow intoxication protocol. The intoxicated animals were classified into asymptomatic, recovered, moderate and severe parkinsonian, based on their motor behavior. The serotonergic innervation was studied by immunohistochemistry against serotonin (5-HT). In the striatum, the density of 5-HT-immunoreactive (5-HT+) axons was estimated with stereology. Images of the striatum in the immunostained sections were taken to compare the distribution patterns of the serotonergic innervation between groups. These patterns were apparently similar among the groups. Axonal density estimations showed no differences in striatal 5-HT+ innervation between the intoxicated groups and the control group. Accordingly, this study fails to find significant changes in the striatal serotonergic axonal innervation in MPTP-treated monkeys, coinciding with previous biochemical findings in our model. However, it is possible that alterations in the serotonergic system in PD could be independent of axonal density changes. Consequently, the proposed role for striatal serotonin serving as a compensatory mechanism for dopaminergic denervation merits further study. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, CEU-San Pablo University, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Natalia L Del Rey
- HM CINAC, Hospital Universitario HM Puerta del Sur, CEU-San Pablo University, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Mariana H G Monje
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; HM CINAC, Hospital Universitario HM Puerta del Sur, CEU-San Pablo University, Madrid, Spain
| | - José A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, CEU-San Pablo University, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Carmen Cavada
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
26
|
Alpár A, Zahola P, Hanics J, Hevesi Z, Korchynska S, Benevento M, Pifl C, Zachar G, Perugini J, Severi I, Leitgeb P, Bakker J, Miklosi AG, Tretiakov E, Keimpema E, Arque G, Tasan RO, Sperk G, Malenczyk K, Máté Z, Erdélyi F, Szabó G, Lubec G, Palkovits M, Giordano A, Hökfelt TG, Romanov RA, Horvath TL, Harkany T. Hypothalamic CNTF volume transmission shapes cortical noradrenergic excitability upon acute stress. EMBO J 2018; 37:e100087. [PMID: 30209240 PMCID: PMC6213283 DOI: 10.15252/embj.2018100087] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023] Open
Abstract
Stress-induced cortical alertness is maintained by a heightened excitability of noradrenergic neurons innervating, notably, the prefrontal cortex. However, neither the signaling axis linking hypothalamic activation to delayed and lasting noradrenergic excitability nor the molecular cascade gating noradrenaline synthesis is defined. Here, we show that hypothalamic corticotropin-releasing hormone-releasing neurons innervate ependymal cells of the 3rd ventricle to induce ciliary neurotrophic factor (CNTF) release for transport through the brain's aqueductal system. CNTF binding to its cognate receptors on norepinephrinergic neurons in the locus coeruleus then initiates sequential phosphorylation of extracellular signal-regulated kinase 1 and tyrosine hydroxylase with the Ca2+-sensor secretagogin ensuring activity dependence in both rodent and human brains. Both CNTF and secretagogin ablation occlude stress-induced cortical norepinephrine synthesis, ensuing neuronal excitation and behavioral stereotypes. Cumulatively, we identify a multimodal pathway that is rate-limited by CNTF volume transmission and poised to directly convert hypothalamic activation into long-lasting cortical excitability following acute stress.
Collapse
Affiliation(s)
- Alán Alpár
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Péter Zahola
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - János Hanics
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Zsófia Hevesi
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Solomiia Korchynska
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gergely Zachar
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Jessica Perugini
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Ilenia Severi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Patrick Leitgeb
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Joanne Bakker
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andras G Miklosi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gloria Arque
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Günther Sperk
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zoltán Máté
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ferenc Erdélyi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gert Lubec
- Paracelsus Medical University, Salzburg, Austria
| | - Miklós Palkovits
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| | - Antonio Giordano
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Tomas Gm Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Departments of Comparative Medicine and Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
27
|
Guridi J, Rodriguez-Rojas R, Carmona-Abellán M, Parras O, Becerra V, Lanciego JL. History and future challenges of the subthalamic nucleus as surgical target: Review article. Mov Disord 2018; 33:1540-1550. [DOI: 10.1002/mds.92] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 01/22/2023] Open
Affiliation(s)
- Jorge Guridi
- Department of Neurosurgery, Neurology and Neurosciences; Clínica Universidad de Navarra, University of Navarra; Pamplona Spain
- Instituto de Investigación Sanitaria Navarra; Pamplona Spain
| | - Rafael Rodriguez-Rojas
- Centro Integral de Neurociencias; University Hospital HM Puerta del Sur; Móstoles Madrid Spain
| | - Mar Carmona-Abellán
- Department of Neurosurgery, Neurology and Neurosciences; Clínica Universidad de Navarra, University of Navarra; Pamplona Spain
- Instituto de Investigación Sanitaria Navarra; Pamplona Spain
| | - Olga Parras
- Department of Neurosurgery, Neurology and Neurosciences; Clínica Universidad de Navarra, University of Navarra; Pamplona Spain
- Instituto de Investigación Sanitaria Navarra; Pamplona Spain
| | - Victoria Becerra
- Department of Neurosurgery, Neurology and Neurosciences; Clínica Universidad de Navarra, University of Navarra; Pamplona Spain
- Instituto de Investigación Sanitaria Navarra; Pamplona Spain
| | - Jose Luis Lanciego
- Department of Neurosurgery, Neurology and Neurosciences; Clínica Universidad de Navarra, University of Navarra; Pamplona Spain
- Instituto de Investigación Sanitaria Navarra; Pamplona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas; Pamplona Spain
| |
Collapse
|
28
|
Choudhury GR, Daadi MM. Charting the onset of Parkinson-like motor and non-motor symptoms in nonhuman primate model of Parkinson's disease. PLoS One 2018; 13:e0202770. [PMID: 30138454 PMCID: PMC6107255 DOI: 10.1371/journal.pone.0202770] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease is a progressive neurodegenerative disease increasingly affecting our aging population. Remarkable advances have been made in developing novel therapies to control symptoms, halt or cure the disease, ranging from physiotherapy and small molecules to cell and gene therapy. This progress was enabled by the existence of reliable animal models. The nonhuman primate model of Parkinson’s disease emulates the cardinal symptoms of the disease, including tremor, rigidity, bradykinesia, postural instability, freezing and cognitive impairment. However, this model is established through the specific loss of midbrain dopaminergic neurons, while our current knowledge reflects the reality of Parkinson’s disease as a multisystem disease. Parkinson’s disease involves both motor and non-motor symptoms, such as sleep disturbance, olfaction, gastrointestinal dysfunctions, depression and cognitive deficits. Some of the non-motor symptoms emerge earlier at the prodromal phase and worsen with disease progression, yet in basic and translational studies, they are rarely considered as endpoints. In this study, we set to characterize an ensemble of less described motor and non-motor dysfunctions in the marmoset MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) model. We provide evidence that this animal model expresses postural head tremor and a progressive worsening of fine motor skills, movement coordination and cognitive abilities over a 6-month period. We report for the first time a non-invasive approach showing detailed analysis of daytime and nighttime sleep and circadian rhythm disturbance remarkably similar to Parkinson’s disease patients. This study describes the incidence of tremors, motor and non-motor dysfunctions in a preclinical model and highlights the need for their consideration in translating effective new therapeutic approaches for Parkinson’s disease.
Collapse
Affiliation(s)
- Gourav R. Choudhury
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Marcel M. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
- Research Imaging Institute, Departments of Radiology, Cell Systems & Anatomy, University of Texas Health at San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
29
|
Abstract
The dentate nucleus (DN) of the cerebellum is the major output nucleus of the cerebellum and is rich in iron. Quantitative susceptibility mapping (QSM) provides better iron-sensitive MRI contrast to delineate the boundary of the DN than either T2-weighted images or susceptibility-weighted images. Prior DN atlases used T2-weighted or susceptibility-weighted images to create DN atlases. Here, we employ QSM images to develop an improved dentate nucleus atlas for use in imaging studies. The DN was segmented in QSM images from 38 healthy volunteers. The resulting DN masks were transformed to a common space and averaged to generate the DN atlas. The center of mass of the left and right sides of the QSM-based DN atlas in the Montreal Neurological Institute space was -13.8, -55.8, and -36.4 mm, and 13.8, -55.7, and -36.4 mm, respectively. The maximal probability and mean probability of the DN atlas with the individually segmented DNs in this cohort were 100 and 39.3%, respectively, in contrast to the maximum probability of approximately 75% and the mean probability of 23.4 to 33.7% with earlier DN atlases. Using QSM, which provides superior iron-sensitive MRI contrast for delineating iron-rich structures, an improved atlas for the dentate nucleus has been generated. The atlas can be applied to investigate the role of the DN in both normal cortico-cerebellar physiology and the variety of disease states in which it is implicated.
Collapse
|
30
|
Gagnon D, Eid L, Coudé D, Whissel C, Di Paolo T, Parent A, Parent M. Evidence for Sprouting of Dopamine and Serotonin Axons in the Pallidum of Parkinsonian Monkeys. Front Neuroanat 2018; 12:38. [PMID: 29867377 PMCID: PMC5963193 DOI: 10.3389/fnana.2018.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022] Open
Abstract
This light and electron microscopie immunohistochemical quantitative study aimed at determining the state of the dopamine (DA) and serotonin (5-HT) innervations of the internal (GPi) and external (GPe) segments of the pallidum in cynomolgus monkeys (Macaca fascicularis) rendered parkinsonian by systemic injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In contrast to the prominent DA denervation of striatum, the GPi in MPTP monkeys was found to be markedly enriched in DA (TH+) axon varicosities. The posterior sensorimotor region of this major output structure of the basal ganglia was about 8 times more intensely innervated in MPTP monkeys (0.71 ± 0.08 × 106 TH+ axon varicosities/mm3) than in controls (0.09 ± 0.01 × 106). MPTP intoxication also induced a two-fold increase in the density of 5-HT (SERT+) axon varicosities in both GPe and GPi. This augmentation was particularly pronounced anteriorly in the so-called associative and limbic pallidal territories. The total length of the labeled pallidal axons was also significantly increased in MPTP monkeys compared to controls, but the number of DA and 5-HT axon varicosities per axon length unit remained the same in the two groups, indicating that the DA and 5-HT pallidal hyperinnervations seen in MPTP monkeys result from axon sprouting rather than from the appearance of newly formed axon varicosities on non-growing axons. At the ultrastructural level, pallidal TH+ and SERT+ axons were morphologically similar in MPTP and controls, and their synaptic incidence was very low suggesting a volumic mode of transmission. Altogether, our data reveal a significant sprouting of DA and 5-HT pallidal afferents in parkinsonian monkeys, the functional significance of which remains to be determined. We suggest that the marked DA hyperinnervation of the GPi represents a neuroadaptive change designed to normalize pallidal firing patterns associated with the delayed appearance of motor symptoms, whereas the 5-HT hyperinnervation might be involved in the early expression of non-motor symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Dave Gagnon
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Lara Eid
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Dymka Coudé
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Carl Whissel
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Thérèse Di Paolo
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
| | - André Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Martin Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
31
|
Emborg ME. Nonhuman Primate Models of Neurodegenerative Disorders. ILAR J 2017; 58:190-201. [PMID: 28985333 PMCID: PMC5886328 DOI: 10.1093/ilar/ilx021] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's (AD), Huntington's (HD), and Parkinson's (PD) disease are age-related neurodegenerative disorders characterized by progressive neuronal cell death. Although each disease has particular pathologies and symptoms, accumulated evidence points to similar mechanisms of neurodegeneration, including inflammation, oxidative stress, and protein aggregation. A significant body of research is ongoing to understand how these pathways affect each other and what ultimately triggers the onset of the disease. Experiments in nonhuman primates (NHPs) account for only 5% of all research in animals. Yet the impact of NHP studies for clinical translation is much greater, especially for neurodegenerative disorders, as NHPs have a complex cognitive and motor functions and highly developed neuroanatomy. New NHP models are emerging to better understand pathology and improve the platform in which to test novel therapies. The goal of this report is to review NHP models of AD, HD, and PD in the context of the current understanding of these diseases and their contribution to the development of novel therapies.
Collapse
Affiliation(s)
- Marina E Emborg
- Marina E. Emborg, MD, PhD, is the director of the Preclinical Parkinson’s Research Program at the Wisconsin National Primate Research Center and an associate professor in the department of Medical Physics at the University of Wisconsin in Madison, Wisconsin.
| |
Collapse
|
32
|
Jo T, Yoshimi K, Takahashi T, Oyama G, Hattori N. Dual use of rectangular and triangular waveforms in voltammetry using a carbon fiber microelectrode to differentiate norepinephrine from dopamine. J Electroanal Chem (Lausanne) 2017. [DOI: 10.1016/j.jelechem.2017.08.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Nishijima H, Ueno T, Funamizu Y, Ueno S, Tomiyama M. Levodopa treatment and dendritic spine pathology. Mov Disord 2017; 33:877-888. [PMID: 28880414 PMCID: PMC6667906 DOI: 10.1002/mds.27172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with the progressive loss of nigrostriatal dopaminergic neurons. Levodopa is the most effective treatment for the motor symptoms of PD. However, chronic oral levodopa treatment can lead to various motor and nonmotor complications because of nonphysiological pulsatile dopaminergic stimulation in the brain. Examinations of autopsy cases with PD have revealed a decreased number of dendritic spines of striatal neurons. Animal models of PD have revealed altered density and morphology of dendritic spines of neurons in various brain regions after dopaminergic denervation or dopaminergic denervation plus levodopa treatment, indicating altered synaptic transmission. Recent studies using rodent models have reported dendritic spine head enlargement in the caudate‐putamen, nucleus accumbens, primary motor cortex, and prefrontal cortex in cases where chronic levodopa treatment following dopaminergic denervation induced dyskinesia‐like abnormal involuntary movement. Hypertrophy of spines results from insertion of alpha‐amino‐2,3‐dihydro‐5‐methyl‐3‐oxo‐4‐isoxazolepropanoic acid receptors into the postsynaptic membrane. Such spine enlargement indicates hypersensitivity of the synapse to excitatory inputs and is compatible with a lack of depotentiation, which is an electrophysiological hallmark of levodopa‐induced dyskinesia found in the corticostriatal synapses of dyskinetic animals and the motor cortex of dyskinetic PD patients. This synaptic plasticity may be one of the mechanisms underlying the priming of levodopa‐induced complications such as levodopa‐induced dyskinesia and dopamine dysregulation syndrome. Drugs that could potentially prevent spine enlargement, such as calcium channel blockers, N‐methyl‐D‐aspartate receptor antagonists, alpha‐amino‐2,3‐dihydro‐5‐methyl‐3‐oxo‐4‐isoxazolepropanoic acid receptor antagonists, and metabotropic glutamate receptor antagonists, are candidates for treatment of levodopa‐induced complications in PD. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Haruo Nishijima
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tatsuya Ueno
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yukihisa Funamizu
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Shinya Ueno
- Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Aomori Prefectural Central Hospital, Aomori, Japan.,Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
34
|
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
|
35
|
Elsworth JD, Brittan MS, Taylor JR, Sladek JR, al-Tikriti MS, Zea-Ponce Y, Innis RB, Redmond DE, Roth RH. Restoration of Dopamine Transporter Density in the Striatum of Fetal Ventral Mesencephalon-Grafted, but not Sham-Grafted, Mptp-Treated Parkinsonian Monkeys. Cell Transplant 2017; 5:315-25. [PMID: 8689042 DOI: 10.1177/096368979600500220] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transplantation of fetal dopamine neurons to the adult striatum potentially offers a means to reverse the striatal dopamine deficiency that characterizes Parkinson's disease. Many investigations in rodents have supported the hope that neural grafting may be a useful treatment for parkinsonism. However, clinical studies have generally produced more modest improvements in motor abnormalities than observed in lower species. It is possible that the number of fetal dopamine neurons that survive transplantation is insufficient to restore dopaminergic innervation of the large human striatum to a level where striking recovery is obtained. In fact, there has been no quantitative study of graft outgrowth to indicate what portion of the dopamine-depleted striatum might be reinner-vated with present techniques. Furthermore, it has been speculated that regeneration of the host dopamine system in response to the implantation surgery may play an important role in the beneficial effects of neural grafting in primates. The present study used nine 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated parkinsonian monkeys to investigate these issues. Sham implantation procedures produced no increase in either dopamine transporter density (measured by quantitative autoradiography) or tissue dopamine concentration (measured by HPLC) in the striatum of MPTP-treated monkeys. In sham-grafted and nonimplanted MPTP-treated monkeys, the striatal dopamine concentration was reduced by 99%, based on analysis of 16 sampled sites in the caudate nucleus and putamen of each monkey. No behavioral recovery was seen in the sham-grafted and nonimplanted MPTP-treated groups. In contrast, transplantation of fetal dopamine neurons to the caudate nucleus or putamen of MPTP-treated monkeys resulted in a significant elevation of dopamine transporter density and dopamine levels in the grafted striatal nucleus. Each grafted MPTP-treated monkey received ventral mesencephalon dopamine neurons from one donor harvested during putative neurogenesis. Donor ventral mesencephalon was divided equally and implanted into six sites either in the caudate nucleus or putamen. One graft site in each monkey was examined by dopamine transporter autoradiography. In sections in which graft fibers were present, a mean of one-third of the volume of the grafted nucleus was occupied by an elevated density of dopamine transporters. This increase in dopamine transporter density was defined to be at least 5-10% of the control density. However, full behavioral recovery was not observed in the grafted MPTP-treated group. These data provide no support for the hypothesis that regeneration of the host dopamine system occurs in response to a sham implantation procedure in severely parkinsonian monkeys. The current study illustrates the power of the applied techniques for delineating the relationship between the level of host dopamine depletion, the extent of graft-induced dopaminergic restoration, and behavioral recovery.
Collapse
Affiliation(s)
- J D Elsworth
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wichmann T, Bergman H, DeLong MR. Basal ganglia, movement disorders and deep brain stimulation: advances made through non-human primate research. J Neural Transm (Vienna) 2017; 125:419-430. [PMID: 28601961 DOI: 10.1007/s00702-017-1736-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/17/2017] [Indexed: 11/30/2022]
Abstract
Studies in non-human primates (NHPs) have led to major advances in our understanding of the function of the basal ganglia and of the pathophysiologic mechanisms of hypokinetic movement disorders such as Parkinson's disease and hyperkinetic disorders such as chorea and dystonia. Since the brains of NHPs are anatomically very close to those of humans, disease states and the effects of medical and surgical approaches, such as deep brain stimulation (DBS), can be more faithfully modeled in NHPs than in other species. According to the current model of the basal ganglia circuitry, which was strongly influenced by studies in NHPs, the basal ganglia are viewed as components of segregated networks that emanate from specific cortical areas, traverse the basal ganglia, and ventral thalamus, and return to the frontal cortex. Based on the presumed functional domains of the different cortical areas involved, these networks are designated as 'motor', 'oculomotor', 'associative' and 'limbic' circuits. The functions of these networks are strongly modulated by the release of dopamine in the striatum. Striatal dopamine release alters the activity of striatal projection neurons which, in turn, influences the (inhibitory) basal ganglia output. In parkinsonism, the loss of striatal dopamine results in the emergence of oscillatory burst patterns of firing of basal ganglia output neurons, increased synchrony of the discharge of neighboring basal ganglia neurons, and an overall increase in basal ganglia output. The relevance of these findings is supported by the demonstration, in NHP models of parkinsonism, of the antiparkinsonian effects of inactivation of the motor circuit at the level of the subthalamic nucleus, one of the major components of the basal ganglia. This finding also contributed strongly to the revival of the use of surgical interventions to treat patients with Parkinson's disease. While ablative procedures were first used for this purpose, they have now been largely replaced by DBS of the subthalamic nucleus or internal pallidal segment. These procedures are not only effective in the treatment of parkinsonism, but also in the treatment of hyperkinetic conditions (such as chorea or dystonia) which result from pathophysiologic changes different from those underlying Parkinson's disease. Thus, these interventions probably do not counteract specific aspects of the pathophysiology of movement disorders, but non-specifically remove the influence of the different types of disruptive basal ganglia output from the relatively intact portions of the motor circuitry downstream from the basal ganglia. Knowledge gained from studies in NHPs remains critical for our understanding of the pathophysiology of movement disorders, of the effects of DBS on brain network activity, and the development of better treatments for patients with movement disorders and other neurologic or psychiatric conditions.
Collapse
Affiliation(s)
- Thomas Wichmann
- Department of Neurology, Emory University, Atlanta, GA, USA. .,Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA.
| | - Hagai Bergman
- Department of Medical Neurobiology (Physiology), Institute of Medical Research Israel-Canada (IMRIC), Jerusalem, Israel.,The Edmond and Lily Safra Center for Brain Research (ELSC), The Hebrew University, Jerusalem, Israel.,Department of Neurosurgery, Hadassah Medical Center, Jerusalem, Israel
| | | |
Collapse
|
37
|
Pifl C, Reither H, Del Rey NLG, Cavada C, Obeso JA, Blesa J. Early Paradoxical Increase of Dopamine: A Neurochemical Study of Olfactory Bulb in Asymptomatic and Symptomatic MPTP Treated Monkeys. Front Neuroanat 2017; 11:46. [PMID: 28611598 PMCID: PMC5447291 DOI: 10.3389/fnana.2017.00046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/12/2017] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with both motor and non-motor manifestations. Hyposmia is one of the early non-motor symptoms, which can precede motor symptoms by several years. The relationship between hyposmia and PD remains elusive. Olfactory bulb (OB) pathology shows an increased number of olfactory dopaminergic cells, protein aggregates and dysfunction of neurotransmitter systems. In this study we examined tissue levels of dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) and their metabolites, of noradrenaline (NA) and of the amino acid neurotransmitters aspartate, glutamate, taurine and γ-aminobutyric acid in OBs of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treated Macaca fascicularis in different stages, including monkeys who were always asymptomatic, monkeys who recovered from mild parkinsonian signs, and monkeys with stable moderate or severe parkinsonism. DA was increased compared to controls, while neither NA and 5-HT nor the amino acid neurotransmitters were significantly changed. Furthermore, DA increased before stable motor deficits appear with +51% in asymptomatic and +96% in recovered monkeys. Unchanged DA metabolites suggest a special metabolic profile of the newly formed DA neurons. Significant correlation of homovanillic acid (HVA) with taurine single values within the four MPTP groups and of aspartate with taurine within the asymptomatic and recovered MPTP groups, but not within the controls suggest interactions in the OB between taurine and the DA system and taurine and the excitatory neurotransmitter triggered by MPTP. This first investigation of OB in various stages after MPTP administration suggests that the DA increase seems to be an early phenomenon, not requiring profound nigrostriatal neurodegeneration or PD symptoms.
Collapse
Affiliation(s)
- Christian Pifl
- Center for Brain Research, Medical University of ViennaVienna, Austria
| | - Harald Reither
- Center for Brain Research, Medical University of ViennaVienna, Austria
| | - Natalia Lopez-Gonzalez Del Rey
- HM CINAC, Hospital Universitario HM Puerta del SurMostoles, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Ministerio de Ciencia e InnovacionMadrid, Spain
| | - Carmen Cavada
- Departamento de Anatomia, Histologia y Neurociencia, Facultad de Medicina, Universidad Autonoma de MadridMadrid, Spain
| | - Jose A Obeso
- HM CINAC, Hospital Universitario HM Puerta del SurMostoles, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Ministerio de Ciencia e InnovacionMadrid, Spain
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del SurMostoles, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Ministerio de Ciencia e InnovacionMadrid, Spain
| |
Collapse
|
38
|
Huot P, Sgambato-Faure V, Fox SH, McCreary AC. Serotonergic Approaches in Parkinson's Disease: Translational Perspectives, an Update. ACS Chem Neurosci 2017; 8:973-986. [PMID: 28460160 DOI: 10.1021/acschemneuro.6b00440] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) has long been seen as a disorder caused by degeneration of the dopaminergic system, leading to the classic motor manifestations of the disease. However, there is now overwhelming evidence that PD is more than a disease merely caused by dopamine depletion. It is well-known that a myriad of other neurotransmitters are affected by the disease process. One such neurotransmitter is serotonin (5-HT). 5-HT has been shown to play a role in several motor and nonmotor manifestations of PD, including tremor, cognition, depression and psychosis. 5-HT also seems to play a critical role in L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia. A breadth of preclinical studies and clinical trials have been conducted that aimed at modulating the 5-HT system in order to alleviate depression, cognitive deficits, psychosis, and dyskinesia. In this Review, we summarize recent advances in the 5-HT field in PD, but with a translational emphasis. We start by presenting a novel nonhuman primate model of PD that presents with dual dopamine and 5-HT lesions. We then present preclinical and clinical data that introduce new concepts, such as the use of biased and partial agonists, as well as molecules recently introduced to the field of PD, such as eltoprazine, pimavanserin, nelotanserin, and SYN-120, to enhance therapeutic benefit while minimizing adverse events, notably on parkinsonian disability.
Collapse
Affiliation(s)
- Philippe Huot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 0A9, Canada
- Department
of Pharmacology, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Unité
des Troubles du Mouvement André Barbeau, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2L 4M1, Canada
- Division
of Neurology, Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Véronique Sgambato-Faure
- Institute of Cognitive
Neuroscience Marc Jeannerod, UMR 5229 CNRS, 69 675 Cedex Bron, France
- University Lyon 1, 69100 Villeurbanne, France
| | - Susan H. Fox
- Movement
Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Andrew C. McCreary
- Janssen Vaccines & Prevention B.V., Archimedesweg 4, 2333 CN Leiden, The Netherlands
| |
Collapse
|
39
|
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
|
40
|
Kanazawa M, Ohba H, Nishiyama S, Kakiuchi T, Tsukada H. Effect of MPTP on Serotonergic Neuronal Systems and Mitochondrial Complex I Activity in the Living Brain: A PET Study on Conscious Rhesus Monkeys. J Nucl Med 2017; 58:1111-1116. [PMID: 28280215 DOI: 10.2967/jnumed.116.189159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
The objective of the present PET study was to compare the effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on serotonergic neuronal systems and mitochondrial complex I (MC-I) activity with that of dopamine in conscious rhesus monkeys (Macaca mulatta). Methods: A Parkinson disease monkey model was prepared by repeated administration of MPTP. For the PET measurements, normal and MPTP-treated conscious monkeys received an intravenous injection of 11C-DASB for serotonin transporter, 18F-MPPF for serotonin 1A receptor, 11C-PE2I for dopamine transporter, 11C-6MemTyr for dopamine synthesis, 11C-raclopride for dopamine D2 receptor, or 18F-BCPP-EF for MC-I. Serotonin and dopamine parameters were calculated using time-activity curves in the cerebellum as the input function. The total distribution volume of 18F-BCPP-EF was assessed using Logan plot graphical analysis with metabolite-corrected plasma as the input function. Results: MPTP-induced diffuse reductions in MC-I activity were observed throughout the brain, except the cerebellum. Significant reductions in the presynaptic dopamine parameters-dopamine transporter and dopamine synthesis-were detected in the striatum and substantia nigra pars compacta of MPTP-treated monkeys, whereas no significant differences in postsynaptic dopamine D2 receptor binding were observed. Serotonin transporter binding was reduced by MPTP not only in striatal regions but also in extrastriatal regions. In contrast, serotonin 1A receptor binding was unaffected by MPTP anywhere in the brain. In the cortex, the reduction of serotonin transporter binding correlated with that of MC-I. Conclusion: The results obtained by multiparametric PET measurements in a Parkinson disease monkey model demonstrated that chronic MPTP treatment induced reductions not only in the dopaminergic system in the nigrostriatal pathway but also in serotonin transporter in the cortical and subcortical regions. These results suggest that the neurotoxicity of MPTP is not exclusive to the nigrostriatal pathway, as predicted from MC-I damage in the extrastriatal regions of the brain.
Collapse
Affiliation(s)
- Masakatsu Kanazawa
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Shizuoka, Japan
| | - Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Shizuoka, Japan
| | - Shingo Nishiyama
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Shizuoka, Japan
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Shizuoka, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Shizuoka, Japan
| |
Collapse
|
41
|
Dopamine and serotonin modulation of motor and non-motor functions of the non-human primate striato-pallidal circuits in normal and pathological states. J Neural Transm (Vienna) 2017; 125:485-500. [PMID: 28176009 DOI: 10.1007/s00702-017-1693-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/30/2017] [Indexed: 10/20/2022]
Abstract
Thanks to the non-human primate (NHP), we have shown that the pharmacological disturbance of the anterior striatum or of external globus pallidus triggers a set of motivation and movement disorders, depending on the functional subterritory involved. One can, therefore, assume that the aberrant activity of the different subterritories of basal ganglia (BG) could lead to different behavioral disorders in neuropsychiatric disorders as Tourette's syndrome and Parkinson's disease. We are now addressing in the NHP the impact of modulating dopamine or serotonin within the BG on behavioral disorders. Indeed, we have shown a prominent role of serotonergic degeneration within the ventral striatum and caudate nucleus in neuropsychiatric symptoms in de novo PD patients. Of note, the serotonergic modulation of these BG regions in the NHP plays also a critical role in the induction or treatment of behavioral disorders. Given that both dopamine and serotonin are targeted to treat neuropsychiatric disorders, we are studying the effects of modulating dopamine and serotonin transporters in the different territories of the striatum, and more particularly within the ventral striatum on decision-making processing at both behavioral and neuronal levels. Finally, we evidence the need to extend the pharmacological approach to the receptors of these two neuromodulator systems as the use of substances targeting receptor subtypes preferentially localized in the associative and limbic territories of BG could be very effective to specifically improve the behavioral disorders in Parkinson's disease, Gilles de la Tourette syndrome but also in several psychiatric disorders such as depression, anxiety, anorexia, or impulse control disorders.
Collapse
|
42
|
|
43
|
Masilamoni GJ, Groover O, Smith Y. Reduced noradrenergic innervation of ventral midbrain dopaminergic cell groups and the subthalamic nucleus in MPTP-treated parkinsonian monkeys. Neurobiol Dis 2016; 100:9-18. [PMID: 28042095 DOI: 10.1016/j.nbd.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/22/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
There is anatomical and functional evidence that ventral midbrain dopaminergic (DA) cell groups and the subthalamic nucleus (STN) receive noradrenergic innervation in rodents, but much less is known about these interactions in primates. Degeneration of NE neurons in the locus coeruleus (LC) and related brainstem NE cell groups is a well-established pathological feature of Parkinson's disease (PD), but the development of such pathology in animal models of PD has been inconsistent across species and laboratories. We recently demonstrated 30-40% neuronal loss in the LC, A5 and A6 NE cell groups of rhesus monkeys rendered parkinsonian by chronic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In this study, we used dopamine-beta-hydroxylase (DβH) immunocytochemistry to assess the impact of this neuronal loss on the number of NE terminal-like varicosities in the substantia nigra pars compacta (SNC), ventral tegmental area (VTA), retrorubral field (RRF) and STN of MPTP-treated parkinsonian monkeys. Our findings reveal that the NE innervation of the ventral midbrain and STN of normal monkeys is heterogeneously distributed being far more extensive in the VTA, RRF and dorsal tier of the SNC than in the ventral SNC and STN. In parkinsonian monkeys, all regions underwent a significant (~50-70%) decrease in NE innervation. At the electron microscopic level, some DβH-positive terminals formed asymmetric axo-dendritic synapses in VTA and STN. These findings demonstrate that the VTA, RRF and SNCd are the main ventral midbrain targets of ascending NE inputs, and that these connections undergo a major break-down in chronically MPTP-treated parkinsonian monkeys. This severe degeneration of the ascending NE system may contribute to the pathophysiology of ventral midbrain and STN neurons in PD.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj 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.
| | - Olivia Groover
- 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.
| | - 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
|
44
|
Sitte HH, Pifl C, Rajput AH, Hörtnagl H, Tong J, Lloyd GK, Kish SJ, Hornykiewicz O. Dopamine and noradrenaline, but not serotonin, in the human claustrum are greatly reduced in patients with Parkinson's disease: possible functional implications. Eur J Neurosci 2016; 45:192-197. [PMID: 27741357 DOI: 10.1111/ejn.13435] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/30/2016] [Accepted: 10/10/2016] [Indexed: 11/28/2022]
Abstract
In the human brain, the claustrum is a small subcortical telencephalic nucleus, situated between the insular cortex and the putamen. A plethora of neuroanatomical studies have shown the existence of dense, widespread, bidirectional and bilateral monosynaptic interconnections between the claustrum and most cortical areas. A rapidly growing body of experimental evidence points to the integrative role of claustrum in complex brain functions, from motor to cognitive. Here, we examined for the first time, the behaviour of the classical monoamine neurotransmitters dopamine, noradrenaline and serotonin in the claustrum of the normal autopsied human brain and of patients who died with idiopathic Parkinson's disease (PD). We found in the normal claustrum substantial amounts of all three monoamine neurotransmitters, substantiating the existence of the respective brain stem afferents to the claustrum. In PD, the levels of dopamine and noradrenaline were greatly reduced by 93 and 81%, respectively. Serotonin levels remained unchanged. We propose that by virtue of their projections to the claustrum, the brain stem dopamine, noradrenaline and serotonin systems interact directly with the cortico-claustro-cortical information processing mechanisms, by-passing their (parallel) routes via the basal ganglia-thalamo-cortical circuits. We suggest that loss of dopamine and noradrenaline in the PD claustrum is critical in the aetiology of both the motor and the non-motor symptoms of PD.
Collapse
Affiliation(s)
- Harald H Sitte
- Centre for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstrasse 13A, A-1090, Vienna, Austria
| | - Christian Pifl
- Centre for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Ali H Rajput
- Division of Neurology, Saskatchewan Movement Disorders Program, Saskatoon Health Region, University of Saskatchewan, Saskatoon, SK, Canada
| | - Heide Hörtnagl
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Junchao Tong
- Human Brain Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | - Stephen J Kish
- Human Brain Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Oleh Hornykiewicz
- Centre for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
De Deurwaerdère P, Di Giovanni G, Millan MJ. Expanding the repertoire of L-DOPA's actions: A comprehensive review of its functional neurochemistry. Prog Neurobiol 2016; 151:57-100. [PMID: 27389773 DOI: 10.1016/j.pneurobio.2016.07.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/18/2016] [Accepted: 07/03/2016] [Indexed: 01/11/2023]
Abstract
Though a multi-facetted disorder, Parkinson's disease is prototypically characterized by neurodegeneration of nigrostriatal dopaminergic neurons of the substantia nigra pars compacta, leading to a severe disruption of motor function. Accordingly, L-DOPA, the metabolic precursor of dopamine (DA), is well-established as a treatment for the motor deficits of Parkinson's disease despite long-term complications such as dyskinesia and psychiatric side-effects. Paradoxically, however, despite the traditional assumption that L-DOPA is transformed in residual striatal dopaminergic neurons into DA, the mechanism of action of L-DOPA is neither simple nor entirely clear. Herein, focussing on its influence upon extracellular DA and other neuromodulators in intact animals and experimental models of Parkinson's disease, we highlight effects other than striatal generation of DA in the functional profile of L-DOPA. While not excluding a minor role for glial cells, L-DOPA is principally transformed into DA in neurons yet, interestingly, with a more important role for serotonergic than dopaminergic projections. Moreover, in addition to the striatum, L-DOPA evokes marked increases in extracellular DA in frontal cortex, nucleus accumbens, the subthalamic nucleus and additional extra-striatal regions. In considering its functional profile, it is also important to bear in mind the marked (probably indirect) influence of L-DOPA upon cholinergic, GABAergic and glutamatergic neurons in the basal ganglia and/or cortex, while anomalous serotonergic transmission is incriminated in the emergence of L-DOPA elicited dyskinesia and psychosis. Finally, L-DOPA may exert intrinsic receptor-mediated actions independently of DA neurotransmission and can be processed into bioactive metabolites. In conclusion, L-DOPA exerts a surprisingly complex pattern of neurochemical effects of much greater scope that mere striatal transformation into DA in spared dopaminergic neurons. Their further experimental and clinical clarification should help improve both L-DOPA-based and novel strategies for controlling the motor and other symptoms of Parkinson's disease.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- CNRS (Centre National de la Recherche Scientifique), Institut des Maladies Neurodégénératives, UMR CNRS 5293, F-33000 Bordeaux, France.
| | - Giuseppe Di Giovanni
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK; Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | - Mark J Millan
- Institut de Recherche Servier, Pole for Therapeutic Innovation in Neuropsychiatry, 78290 Croissy/Seine,Paris, France
| |
Collapse
|
46
|
Peneder TM, Bauer J, Pifl C. Apoptosis-inducing factor in nigral dopamine neurons: Higher levels in primates than in mice. Mov Disord 2016; 31:1729-1733. [PMID: 27297192 DOI: 10.1002/mds.26695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION The nigrostriatal dopaminergic pathway is more susceptible to neurodegeneration in primates than in mice, including the neurotoxic effect of MPTP. Apoptosis-inducing-factor was shown to be involved in the pathogenesis of dopaminergic degeneration. We therefore compared its occurrence in nigral dopamine neurons of mice, monkeys, and humans. METHODS Paraffin-embedded brain slices, including the SNpc of C57BL/6J mice, rhesus monkeys, and humans, were immunohistochemically labeled for tyrosine hydroxylase (an enzyme of dopamine synthesis), microtubule-associated protein 2 (a neuronal marker), and apoptosis-inducing factor and examined by confocal laser scan microscopy. RESULTS The amount of apoptosis-inducing factor in TH-containing SN neurons was 15 times higher in monkeys and 50 times higher in humans than in mice in terms of apoptosis-inducing factor immunoreactive neuronal area excluding the nucleus. CONCLUSION The difference of apoptosis-inducing factor levels between primates and mice might contribute to the higher sensitivity of primates to MPTP-induced neurodegeneration of their nigrostriatal dopamine system. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tamara M Peneder
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
De Deurwaerdère P, Di Giovanni G. Serotonergic modulation of the activity of mesencephalic dopaminergic systems: Therapeutic implications. Prog Neurobiol 2016; 151:175-236. [PMID: 27013075 DOI: 10.1016/j.pneurobio.2016.03.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
Abstract
Since their discovery in the mammalian brain, it has been apparent that serotonin (5-HT) and dopamine (DA) interactions play a key role in normal and abnormal behavior. Therefore, disclosure of this interaction could reveal important insights into the pathogenesis of various neuropsychiatric diseases including schizophrenia, depression and drug addiction or neurological conditions such as Parkinson's disease and Tourette's syndrome. Unfortunately, this interaction remains difficult to study for many reasons, including the rich and widespread innervations of 5-HT and DA in the brain, the plethora of 5-HT receptors and the release of co-transmitters by 5-HT and DA neurons. The purpose of this review is to present electrophysiological and biochemical data showing that endogenous 5-HT and pharmacological 5-HT ligands modify the mesencephalic DA systems' activity. 5-HT receptors may control DA neuron activity in a state-dependent and region-dependent manner. 5-HT controls the activity of DA neurons in a phasic and excitatory manner, except for the control exerted by 5-HT2C receptors which appears to also be tonically and/or constitutively inhibitory. The functional interaction between the two monoamines will also be discussed in view of the mechanism of action of antidepressants, antipsychotics, anti-Parkinsonians and drugs of abuse.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5293, 33076 Bordeaux Cedex, France.
| | - Giuseppe Di Giovanni
- Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
48
|
Franke SK, van Kesteren RE, Hofman S, Wubben JAM, Smit AB, Philippens IHCHM. Individual and Familial Susceptibility to MPTP in a Common Marmoset Model for Parkinson's Disease. NEURODEGENER DIS 2016; 16:293-303. [PMID: 26999593 DOI: 10.1159/000442574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Insight into susceptibility mechanisms underlying Parkinson's disease (PD) would aid the understanding of disease etiology, enable target finding and benefit the development of more refined disease-modifying strategies. METHODS We used intermittent low-dose MPTP (0.5 mg/kg/week) injections in marmosets and measured multiple behavioral and neurochemical parameters. Genetically diverse monkeys from different breeding families were selected to investigate inter- and intrafamily differences in susceptibility to MPTP treatment. RESULTS We show that such differences exist in clinical signs, in particular nonmotor PD-related behaviors, and that they are accompanied by differences in neurotransmitter levels. In line with the contribution of a genetic component, different susceptibility phenotypes could be traced back through genealogy to individuals of the different families. CONCLUSION Our findings show that low-dose MPTP treatment in marmosets represents a clinically relevant PD model, with a window of opportunity to examine the onset of the disease, allowing the detection of individual variability in disease susceptibility, which may be of relevance for the diagnosis and treatment of PD in humans.
Collapse
Affiliation(s)
- Sigrid K Franke
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
49
|
Gagnon D, Gregoire L, Di Paolo T, Parent M. Serotonin hyperinnervation of the striatum with high synaptic incidence in parkinsonian monkeys. Brain Struct Funct 2015; 221:3675-91. [DOI: 10.1007/s00429-015-1125-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/29/2015] [Indexed: 12/23/2022]
|
50
|
Zhang X, Song D, Gu L, Ren Y, Verkhratsky A, Peng L. Decrease of gene expression of astrocytic 5-HT2B receptors parallels development of depressive phenotype in a mouse model of Parkinson's disease. Front Cell Neurosci 2015; 9:388. [PMID: 26500493 PMCID: PMC4594497 DOI: 10.3389/fncel.2015.00388] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/17/2015] [Indexed: 12/12/2022] Open
Abstract
Astrocytes contribute to pathogenesis of neuropsychiatric disorders, including major depression. Stimulation of astroglial 5-HT2B receptors transactivates epidermal growth factor receptors (EGFRs) and regulates gene expression. Previously we reported that expression of 5-HT2B receptors in cortical astrocytes is down-regulated in animals, which developed anhedonia in response to chronic stress; moreover this down-regulation as well as anhedonia, are reversed by chronic treatment with fluoxetine. In this study we have investigated whether astrocytic 5-HT2B receptor is involved in anhedonia in C57BL/6 mice model of Parkinson' disease (PD) induced by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) for 7 days. The MPTP treatment induced anhendonia in 66.7% of animals. The appearance of depressive behavior was accompanied with motor deficiency and decrease of tyrosine hydroxylase (TH) expression. Expression of mRNA and protein of 5-HT2B receptor in animals that became anhedonic decreased to 77.3 and 79.3% of control groups, respectively; in animals that received MPTP but did not develop anhedonia the expression of 5-HT2B receptor did not change. Experiments with FACS-sorted isolated cells demonstrated that decrease in 5-HT2B receptor expression was confined to astrocytes, and did not occur in neurons. Fluoxetine corrected MPTP-induced decrease of 5-HT2B receptor expression and depressive behavior. Our findings indicate that regulation of gene expression of 5-HT2B receptors in astroglia may be associated with pathophysiological evolution of PD-induced depression.
Collapse
Affiliation(s)
- Xique Zhang
- Department of Neurology, The First Affiliated Hospital, China Medical University Shenyang, China
| | - Dan Song
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| | - Li Gu
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| | - Yan Ren
- Department of Neurology, The First Affiliated Hospital, China Medical University Shenyang, China
| | - Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester Manchester, UK ; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science Bilbao, Spain ; University of Nizhny Novgorod Nizhny Novgorod, Russia
| | - Liang Peng
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| |
Collapse
|