1
|
Jiang X, Sun M, Yan Y, Wang Y, Fan X, Wei J, Wang K, Liang P, Wang Z, Wang J, Wang X, Jia J. Corticostriatal glutamate-mediated dynamic therapeutic efficacy of electroacupuncture in a parkinsonian rat model. Clin Transl Med 2024; 14:e70117. [PMID: 39627032 PMCID: PMC11614550 DOI: 10.1002/ctm2.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Motor impairments are the defining cardinal features of Parkinson's disease (PD), resulting from malfunction of the cortico-basal ganglia circuit. Clinical data have demonstrated that electroacupuncture (EA) stimulation may benefit motor symptoms in PD without adverse effects. However, the specific effects of EA on PD and the underlying mechanisms remain largely unclear. METHODS This study investigated the effects of EA stimulation during and after 100 Hz application in a rat model of PD created by unilateral injection of 6-hydroxydopamine (6-OHDA). To establish optimal treatment parameters of EA, motor behaviours were dynamically assessed using open field and rotarod tests. Additionally, we evaluated corticostriatal spine plasticity using immunoelectron microscopy and measured the levels of dopaminergic and glutamatergic neurotransmitters through microdialysis, in vivo electrochemistry and high-performance liquid chromatography. Neural activity dynamics were recorded by measuring local field potentials in both the motor cortex and the striatum. Furthermore, chemogenetic techniques were employed to manipulate corticostriatal glutamatergic neurons and clarify the mechanisms that contribute to the therapeutic benefits of EA in the PD rat model. RESULTS Chronic EA stimulation resulted in a gradual and long-lasting alleviation of motor symptoms, independent of nigrostriatal dopamine (DA) restoration. Notably, EA stimulation modulated corticostriatal spine plasticity and reduced excessive glutamate transmission in PD model rats. Moreover, EA effectively inhibited aberrant corticostriatal synchronised high-beta (25-40 Hz) oscillations, which serves as a pathological biomarker of PD. Conversely, chronic chemogenetic activation of corticostriatal glutamatergic neurons hindered these positive outcomes of EA treatment in PD model rats. CONCLUSIONS This study sheds light on the temporal dynamics and optimal parameters of EA treatment in PD. It emphasises the significance of inhibiting corticostriatal glutamate transmission in EA's therapeutic benefits for PD. Targeting glutamatergic neurons with EA holds promise as a non-dopaminergic intervention for managing motor symptoms and abnormal neural activity with PD. KEY POINTS EA commonly protects dopaminergic neuronsby reducing neuroinflammation, oxidative stress, and apoptosis. New findings reveal that EA alleviates motor symptoms in a parkinsonian rat model without restoring striatal dopamine levels. EA effectively suppresses excessiveglutamate transmission and high-beta synchronization, contributing to motorsymptom relief. Activation of corticostriatalglutamatergic projections may hinder the efficacy of EA.
Collapse
Affiliation(s)
- Xinxin Jiang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Min Sun
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
- Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yitong Yan
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Yanhua Wang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Xinyu Fan
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jing Wei
- School of Biomedical EngineeringCapital Medical UniversityBeijingChina
| | - Ke Wang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Peirong Liang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Zirui Wang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jihan Wang
- School of Biomedical EngineeringCapital Medical UniversityBeijingChina
| | - Xiaomin Wang
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jun Jia
- Department of Physiology and PathophysiologySchool of Basic Medical ScienceCapital Medical UniversityBeijingChina
| |
Collapse
|
2
|
Amato S, Averna M, Farsetti E, Guidolin D, Pedrazzi M, Gatta E, Candiani S, Maura G, Agnati LF, Cervetto C, Marcoli M. Control of Dopamine Signal in High-Order Receptor Complex on Striatal Astrocytes. Int J Mol Sci 2024; 25:8610. [PMID: 39201299 PMCID: PMC11354247 DOI: 10.3390/ijms25168610] [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: 06/28/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The receptor-receptor interaction (RRI) of G protein-coupled receptors (GPCRs) leads to new functional entities that are conceptually distinct from the simple addition of signals mediated by the activation of the receptors that form the heteromers. Focusing on astrocytes, there is evidence for the existence of inhibitory and facilitatory RRIs, including the heteromers formed by the adenosine A2A and the dopamine D2 receptors, by A2A and the oxytocin receptor (OTR), and the D2-OTR heteromers. The possible involvement of these receptors in mosaicism has never been investigated in striatal astrocytes. By biophysical and functional approaches, we focused our attention on the existence of an A2A-D2-OTR high-order receptor complex and its role in modulating cytosolic calcium levels and endogenous glutamate release, when striatal astrocyte processes were stimulated with 4-aminopyridine. Functional data indicate a permissive role of OTR on dopamine signaling in the regulation of the glutamatergic transmission, and an inhibitory control mediated by A2A on both the D2-mediated signaling and on the OTR-facilitating effect on D2. Imaging biochemical and bioinformatic evidence confirmed the existence of the A2A-D2-OTR complex and its ternary structure in the membrane. In conclusion, the D2 receptor appears to be a hotspot in the control of the glutamate release from the astrocytic processes and may contribute to the regulation and integration of different neurotransmitter-mediated signaling in the striatum by the A2A-D2-OTR heterotrimers. Considering the possible selectivity of allosteric interventions on GPCRs organized as receptor mosaics, A2A-D2-OTR heterotrimers may offer selective pharmacological targets in neuropsychiatric disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Monica Averna
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elisa Farsetti
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Via Gabelli 63, 35122 Padova, Italy
| | - Marco Pedrazzi
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elena Gatta
- DIFILAB, Department of Physics, University of Genova, Via Dodecaneso 33, 16146 Genova, Italy
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Via Largo Benzi 10, 16132 Genova, Italy
| | - Guido Maura
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
| | - Luigi Francesco Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Via Largo Benzi 10, 16132 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Manuela Marcoli
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
3
|
Masilamoni GJ, Kelly H, Swain AJ, Pare JF, Villalba RM, Smith Y. Structural Plasticity of GABAergic Pallidothalamic Terminals in MPTP-Treated Parkinsonian Monkeys: A 3D Electron Microscopic Analysis. eNeuro 2024; 11:ENEURO.0241-23.2024. [PMID: 38514185 PMCID: PMC10957232 DOI: 10.1523/eneuro.0241-23.2024] [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: 07/09/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
The internal globus pallidus (GPi) is a major source of tonic GABAergic inhibition to the motor thalamus. In parkinsonism, the firing rate of GPi neurons is increased, and their pattern switches from a tonic to a burst mode, two pathophysiological changes associated with increased GABAergic pallidothalamic activity. In this study, we used high-resolution 3D electron microscopy to demonstrate that GPi terminals in the parvocellular ventral anterior nucleus (VApc) and the centromedian nucleus (CM), the two main GPi-recipient motor thalamic nuclei in monkeys, undergo significant morphometric changes in parkinsonian monkeys including (1) increased terminal volume in both nuclei; (2) increased surface area of synapses in both nuclei; (3) increased number of synapses/GPi terminals in the CM, but not VApc; and (4) increased total volume, but not number, of mitochondria/terminals in both nuclei. In contrast to GPi terminals, the ultrastructure of putative GABAergic nonpallidal terminals was not affected. Our results also revealed striking morphological differences in terminal volume, number/area of synapses, and volume/number of mitochondria between GPi terminals in VApc and CM of control monkeys. In conclusion, GABAergic pallidothalamic terminals are endowed with a high level of structural plasticity that may contribute to the development and maintenance of the abnormal increase in pallidal GABAergic outflow to the thalamus in the parkinsonian state. Furthermore, the evidence for ultrastructural differences between GPi terminals in VApc and CM suggests that morphologically distinct pallidothalamic terminals from single pallidal neurons may underlie specific physiological properties of pallidal inputs to VApc and CM in normal and diseased states.
Collapse
Affiliation(s)
- G J Masilamoni
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
| | - H Kelly
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
| | - A J Swain
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
| | - J F Pare
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
| | - R M Villalba
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
| | - Y Smith
- Emory National Primate Research Center, Atlanta, Georgia 30322
- Udall Center of Excellence for Parkinson's Disease, Emory University, Atlanta, Georgia 30322
- Department of Neurology, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
4
|
Chaudhary R, Singh R. Therapeutic Viewpoint on Rat Models of Locomotion Abnormalities and Neurobiological Indicators in Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:488-503. [PMID: 37202886 DOI: 10.2174/1871527322666230518111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 05/20/2023]
Abstract
BACKGROUND Locomotion problems in Parkinson's syndrome are still a research and treatment difficulty. With the recent introduction of brain stimulation or neuromodulation equipment that is sufficient to monitor activity in the brain using electrodes placed on the scalp, new locomotion investigations in patients having the capacity to move freely have sprung up. OBJECTIVE This study aimed to find rat models and locomotion-connected neuronal indicators and use them all over a closed-loop system to enhance the future and present treatment options available for Parkinson's disease. METHODS Various publications on locomotor abnormalities, Parkinson's disease, animal models, and other topics have been searched using several search engines, such as Google Scholar, Web of Science, Research Gate, and PubMed. RESULTS Based on the literature, we can conclude that animal models are used for further investigating the locomotion connectivity deficiencies of many biological measuring devices and attempting to address unanswered concerns from clinical and non-clinical research. However, translational validity is required for rat models to contribute to the improvement of upcoming neurostimulation-based medicines. This review discusses the most successful methods for modelling Parkinson's locomotion in rats. CONCLUSION This review article has examined how scientific clinical experiments lead to localised central nervous system injuries in rats, as well as how the associated motor deficits and connection oscillations reflect this. This evolutionary process of therapeutic interventions may help to improve locomotion- based treatment and management of Parkinson's syndrome in the upcoming years.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
- Department of Pharmacology, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
| |
Collapse
|
5
|
Cervetto C, Maura G, Guidolin D, Amato S, Ceccoli C, Agnati LF, Marcoli M. Striatal astrocytic A2A-D2 receptor-receptor interactions and their role in neuropsychiatric disorders. Neuropharmacology 2023:109636. [PMID: 37321323 DOI: 10.1016/j.neuropharm.2023.109636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Italy.
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Cristina Ceccoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Luigi F Agnati
- Department of Biochemical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy; Center of Excellence for Biomedical Research, University of Genova, Italy.
| |
Collapse
|
6
|
Heteromerization of Dopamine D2 and Oxytocin Receptor in Adult Striatal Astrocytes. Int J Mol Sci 2023; 24:ijms24054677. [PMID: 36902106 PMCID: PMC10002782 DOI: 10.3390/ijms24054677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
The ability of oxytocin (OT) to interact with the dopaminergic system through facilitatory D2-OT receptor (OTR) receptor-receptor interaction in the limbic system is increasingly considered to play roles in social or emotional behavior, and suggested to serve as a potential therapeutic target. Although roles of astrocytes in the modulatory effects of OT and dopamine in the central nervous system are well recognized, the possibility of D2-OTR receptor-receptor interaction in astrocytes has been neglected. In purified astrocyte processes from adult rat striatum, we assessed OTR and dopamine D2 receptor expression by confocal analysis. The effects of activation of these receptors were evaluated in the processes through a neurochemical study of glutamate release evoked by 4-aminopyridine; D2-OTR heteromerization was assessed by co-immunoprecipitation and proximity ligation assay (PLA). The structure of the possible D2-OTR heterodimer was estimated by a bioinformatic approach. We found that both D2 and OTR were expressed on the same astrocyte processes and controlled the release of glutamate, showing a facilitatory receptor-receptor interaction in the D2-OTR heteromers. Biochemical and biophysical evidence confirmed D2-OTR heterodimers on striatal astrocytes. The residues in the transmembrane domains four and five of both receptors are predicted to be mainly involved in the heteromerization. In conclusion, roles for astrocytic D2-OTR in the control of glutamatergic synapse functioning through modulation of astrocytic glutamate release should be taken into consideration when considering interactions between oxytocinergic and dopaminergic systems in striatum.
Collapse
|
7
|
Xylaki M, Paiva I, Al-Azzani M, Gerhardt E, Jain G, Islam MR, Vasili E, Wassouf Z, Schulze-Hentrich JM, Fischer A, Outeiro TF. miR-101a-3p Impairs Synaptic Plasticity and Contributes to Synucleinopathy. JOURNAL OF PARKINSON'S DISEASE 2023; 13:179-196. [PMID: 36744345 PMCID: PMC10041420 DOI: 10.3233/jpd-225055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Synucleinopathies are disorders characterized by the abnormal accumulation of α-synuclein (aSyn). Synaptic compromise is observed in synucleinopathies parallel to aSyn aggregation and is accompanied by transcript deregulation. OBJECTIVE We sought to identify microRNAs associated with synaptic processes that may contribute to synaptic dysfunction and degeneration in synucleinopathies. METHODS We performed small RNA-sequencing of midbrain from 6-month-old transgenic mice expressing A30P mutant aSyn, followed by comparative expression analysis. We then used real-time quantitative polymerase chain reaction (qPCR) for validation. Functional analysis was performed in primary neurons by biochemical assays and imaging. RESULTS We found several deregulated biological processes linked to the synapse. miR-101a-3p was validated as a synaptic miRNA upregulated in aSyn Tg mice and in the cortex of dementia with Lewy bodies patients. Mice and primary cultured neurons overexpressing miR-101a-3p showed downregulation of postsynaptic proteins GABA Ab2 and SAPAP3 and altered dendritic morphology resembling synaptic plasticity impairments and/or synaptic damage. Interestingly, primary cultured neuron exposure to recombinant wild-type aSyn species efficiently increased miR-101a-3p levels. Finally, a dynamic role of miR-101a-3p in synapse plasticity was shown by identifying downregulation of miR-101a-3p in a condition of enhanced synaptic plasticity modelled in Wt animals housed in enriched environment. CONCLUSION To conclude, we correlated pathologic aSyn with high levels of miR-101a-3p and a novel dynamic role of the miRNA in synaptic plasticity.
Collapse
Affiliation(s)
- Mary Xylaki
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Isabel Paiva
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
- Present address: Laboratory of Cognitive and Adaptive Neuroscience, UMR 7364 (CNRS/ Strasbourg University), Strasbourg, France
| | - Mohammed Al-Azzani
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Ellen Gerhardt
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Zinah Wassouf
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | | | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific employee with an honorary contract at German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
8
|
Gonçalves FQ, Matheus FC, Silva HB, Real JI, Rial D, Rodrigues RJ, Oses JP, Silva AC, Gonçalves N, Prediger RD, Tomé ÂR, Cunha RA. Increased ATP Release and Higher Impact of Adenosine A 2A Receptors on Corticostriatal Plasticity in a Rat Model of Presymptomatic Parkinson's Disease. Mol Neurobiol 2023; 60:1659-1674. [PMID: 36547848 PMCID: PMC9899190 DOI: 10.1007/s12035-022-03162-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Extracellular ATP can be a danger signal, but its role in striatal circuits afflicted in Parkinson's disease (PD) is unclear and was now investigated. ATP was particularly released at high stimulation intensities from purified striatal nerve terminals of mice, which were endowed with different ATP-P2 receptors (P2R), although P2R antagonists did not alter corticostriatal transmission or plasticity. Instead, ATP was extracellularly catabolized into adenosine through CD73 to activate adenosine A2A receptors (A2AR) modulating corticostriatal long-term potentiation (LTP) in mice. In the presymptomatic phase of a 6-hydroxydopamine rat model of PD, ATP release from striatal nerve terminals was increased and was responsible for a greater impact of CD73 and A2AR on corticostriatal LTP. These observations identify increased ATP release and ATP-derived formation of extracellular adenosine bolstering A2AR activation as a key pathway responsible for abnormal synaptic plasticity in circuits involved in the onset of PD motor symptoms. The translation of these findings to humans prompts extending the use of A2AR antagonists from only co-adjuvants of motor control in Parkinsonian patients to neuroprotective drugs delaying the onset of motor symptoms.
Collapse
Affiliation(s)
| | - Filipe C. Matheus
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC Brazil
| | - Henrique B. Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Joana I. Real
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Daniel Rial
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ricardo J. Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Jean-Pierre Oses
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - António C. Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Nélio Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rui D. Prediger
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC Brazil
| | - Ângelo R. Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
9
|
Fu X, He Y, Xie Y, Lu Z. A conjoint analysis of bulk RNA-seq and single-nucleus RNA-seq for revealing the role of ferroptosis and iron metabolism in ALS. Front Neurosci 2023; 17:1113216. [PMID: 36937665 PMCID: PMC10017473 DOI: 10.3389/fnins.2023.1113216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive and selective degeneration of motor neurons in the motor cortex of brain and spinal cord. Ferroptosis is a newly discovered form of cell death and reported to mediate selective motor neuron death in the mouse model of ALS. The growing awareness of ferroptosis and iron metabolism dysfunction in ALS prompted us to investigate the expression pattern of ferroptosis and iron metabolism-related genes (FIRGs) in ALS. Here, we performed a conjoint analysis of bulk-RNA sequence and single-nucleus RNA sequence data using the datasets from Gene Expression Omnibus (GEO) to reveal the role of FIRGs in ALS, especially in selective motor neuron death of ALS. We first investigated the differentially expressed genes (DEGs) between ALS and non-neurological controls. Weighted gene co-expression network analysis constructed the gene co-expression network and identified three modules closely associated with ALS. Fifteen FIRGs was identified as target genes based on least absolute shrinkage and selection operator regression analysis as follows: ACSL4, ANO6, ATP6V0E1, B2M, CD44, CHMP5, CYBB, CYBRD1, HIF1A, MOSPD1, NCF2, SDCBP, STEAP2, TMEM14C, ULK1. These genes could differentiate ALS patients from non-neurological controls (p < 2.2e-16) and had a valid value in predicting and diagnosing ALS (AUC = 0.881 in primary dataset and AUC = 0.768 in validation dataset). Then we performed the functional enrichment analysis of DEGs between ALS cases, the most significantly influenced by target genes, and non-neurological controls. The result indicated that the most significantly influenced functions in ALS pathogenesis by these identified FIRGs are synapse pathways, calcium signaling pathway, cAMP signaling pathway, and phagosome and several immune pathways. At last, the analysis of single- nuclear seq found that CHMP5, one of the 15 FIRGs identified by bulk single-nucleus RNA-seq data, was expressed significantly higher in ALS than pathologically normal (PN), specifically in excitatory neuron populations with layer 2 and layer 3 markers (Ex L2_L3), layer 3 and layer 5 markers (Ex L3_L5). Taken together, our study indicates the positive correlation between FIRGs and ALS, presents potential markers for ALS diagnosis and provides new research directions of CHMP5 function in selective motor neuron death in ALS.
Collapse
Affiliation(s)
- Xiujuan Fu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yizi He
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yongzhi Xie,
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Zuneng Lu,
| |
Collapse
|
10
|
Padilla-Orozco M, Duhne M, Fuentes-Serrano A, Ortega A, Galarraga E, Bargas J, Lara-González E. Synaptic determinants of cholinergic interneurons hyperactivity during parkinsonism. Front Synaptic Neurosci 2022; 14:945816. [PMID: 36147730 PMCID: PMC9485566 DOI: 10.3389/fnsyn.2022.945816] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/21/2022] [Indexed: 11/25/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative ailment generated by the loss of dopamine in the basal ganglia, mainly in the striatum. The disease courses with increased striatal levels of acetylcholine, disrupting the balance among these modulatory transmitters. These modifications disturb the excitatory and inhibitory balance in the striatal circuitry, as reflected in the activity of projection striatal neurons. In addition, changes in the firing pattern of striatal tonically active interneurons during the disease, including cholinergic interneurons (CINs), are being searched. Dopamine-depleted striatal circuits exhibit pathological hyperactivity as compared to controls. One aim of this study was to show how striatal CINs contribute to this hyperactivity. A second aim was to show the contribution of extrinsic synaptic inputs to striatal CINs hyperactivity. Electrophysiological and calcium imaging recordings in Cre-mice allowed us to evaluate the activity of dozens of identified CINs with single-cell resolution in ex vivo brain slices. CINs show hyperactivity with bursts and silences in the dopamine-depleted striatum. We confirmed that the intrinsic differences between the activity of control and dopamine-depleted CINs are one source of their hyperactivity. We also show that a great part of this hyperactivity and firing pattern change is a product of extrinsic synaptic inputs, targeting CINs. Both glutamatergic and GABAergic inputs are essential to sustain hyperactivity. In addition, cholinergic transmission through nicotinic receptors also participates, suggesting that the joint activity of CINs drives the phenomenon; since striatal CINs express nicotinic receptors, not expressed in striatal projection neurons. Therefore, CINs hyperactivity is the result of changes in intrinsic properties and excitatory and inhibitory inputs, in addition to the modification of local circuitry due to cholinergic nicotinic transmission. We conclude that CINs are the main drivers of the pathological hyperactivity present in the striatum that is depleted of dopamine, and this is, in part, a result of extrinsic synaptic inputs. These results show that CINs may be a main therapeutic target to treat Parkinson’s disease by intervening in their synaptic inputs.
Collapse
Affiliation(s)
- Montserrat Padilla-Orozco
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mariana Duhne
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Alejandra Fuentes-Serrano
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aidán Ortega
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elvira Galarraga
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Bargas
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: José Bargas,
| | - Esther Lara-González
- División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Esther Lara-González,
| |
Collapse
|
11
|
Glutamate cycle changes in the putamen of patients with de novo Parkinson's disease using 1H MRS. Parkinsonism Relat Disord 2022; 99:65-72. [PMID: 35613535 DOI: 10.1016/j.parkreldis.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022]
Abstract
INTRODUCTION To investigate glutamatergic metabolism changes in the putamen of patients with de novo Parkinson's Disease (PD) and test the hypothesis that glutamate (Glu) levels are abnormally elevated in the putamen contralateral to where the motor clinical signs predominate as expected from observations in animal models. METHODS 1H NMR spectra from 17 healthy control volunteers were compared with spectra from 17 de novo PD patients of who 14 were evaluated again after 2-3 years of disease progression. Statistical analysis used random-effects models. RESULTS The only significant difference between PD patients and controls was a higher glutamine (Gln) concentration in the putamen ipsilateral to the hemibody with predominant motor signs (Visit 1: 6.0 ± 0.4 mM vs. 5.2 ± 0.2 mM, p < 0.05; Visit 2: 6.2 ± 0.3 mM vs. 5.2 ± 0.2 mM, p < 0.05). At Visit 1, PD patients had higher Glu and Gln levels in the putamen ipsilateral versus contralateral to dominant clinical signs (Glu: 12.2 ± 0.6 mM vs. 10.4 ± 0.6 mM, p < 0.05; Gln: 6.0 ± 0.4 mM vs. 4.8 ± 0.4 mM, p < 0.05; Glu and Gln pool (Glx): 17.9 ± 0.8 mM vs. 14.7 ± 1.1 mM, p < 0.05). At Visit 2, the sum of the two metabolites remained significantly higher in the ipsilateral versus contralateral putamen (Glx: 18.3 ± 0.6 mM vs. 16.1 ± 0.9 mM, p < 0.05). CONCLUSION In de novo PD patients, the putamen ipsilateral to the more affected hemibody showed elevated Gln versus controls and elevated Glu and Gln concentrations versus the contralateral side. Abnormalities in Glu metabolism therefore occur early in PD but unexpectedly in the putamen contralateral to the more damaged hemisphere, suggesting they are not dependent solely on dopamine loss.
Collapse
|
12
|
Merino-Galan L, Jimenez-Urbieta H, Zamarbide M, Rodríguez-Chinchilla T, Belloso-Iguerategui A, Santamaria E, Fernández-Irigoyen J, Aiastui A, Doudnikoff E, Bézard E, Ouro A, Knafo S, Gago B, Quiroga-Varela A, Rodríguez-Oroz MC. Striatal synaptic bioenergetic and autophagic decline in premotor experimental parkinsonism. Brain 2022; 145:2092-2107. [PMID: 35245368 PMCID: PMC9460676 DOI: 10.1093/brain/awac087] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/31/2022] [Accepted: 02/20/2022] [Indexed: 12/02/2022] Open
Abstract
Synaptic impairment might precede neuronal degeneration in Parkinson’s disease. However, the intimate mechanisms altering synaptic function by the accumulation of presynaptic α-synuclein in striatal dopaminergic terminals before dopaminergic death occurs, have not been elucidated. Our aim is to unravel the sequence of synaptic functional and structural changes preceding symptomatic dopaminergic cell death. As such, we evaluated the temporal sequence of functional and structural changes at striatal synapses before parkinsonian motor features appear in a rat model of progressive dopaminergic death induced by overexpression of the human mutated A53T α-synuclein in the substantia nigra pars compacta, a protein transported to these synapses. Sequential window acquisition of all theoretical mass spectra proteomics identified deregulated proteins involved first in energy metabolism and later, in vesicle cycling and autophagy. After protein deregulation and when α-synuclein accumulated at striatal synapses, alterations to mitochondrial bioenergetics were observed using a Seahorse XF96 analyser. Sustained dysfunctional mitochondrial bioenergetics was followed by a decrease in the number of dopaminergic terminals, morphological and ultrastructural alterations, and an abnormal accumulation of autophagic/endocytic vesicles inside the remaining dopaminergic fibres was evident by electron microscopy. The total mitochondrial population remained unchanged whereas the number of ultrastructurally damaged mitochondria increases as the pathological process evolved. We also observed ultrastructural signs of plasticity within glutamatergic synapses before the expression of motor abnormalities, such as a reduction in axospinous synapses and an increase in perforated postsynaptic densities. Overall, we found that a synaptic energetic failure and accumulation of dysfunctional organelles occur sequentially at the dopaminergic terminals as the earliest events preceding structural changes and cell death. We also identify key proteins involved in these earliest functional abnormalities that may be modulated and serve as therapeutic targets to counterbalance the degeneration of dopaminergic cells to delay or prevent the development of Parkinson’s disease.
Collapse
Affiliation(s)
- Leyre Merino-Galan
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Neuroscience Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Haritz Jimenez-Urbieta
- Cell culture Platform, Biodonostia Health Research Institute, San Sebastian, 20014 Donostia, Spain
| | - Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
| | | | | | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ana Aiastui
- Cell culture Platform, Biodonostia Health Research Institute, San Sebastian, 20014 Donostia, Spain
| | - Evelyne Doudnikoff
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Erwan Bézard
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Alberto Ouro
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Shira Knafo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, 8410501 Beer-Sheva, Israel.,Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Basque Foundation for Science, IKERBASQUE, 48940 Leioa, Spain
| | - Belén Gago
- Faculty of Medicine, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29016 Málaga, Spain
| | - Ana Quiroga-Varela
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - María Cruz Rodríguez-Oroz
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Neurology Department, Clínica Universidad de Navarra (CUN), 31008 Pamplona, Spain
| |
Collapse
|
13
|
Voronkov D, Stavrovskaya A, Olshanskiy A, Guschina A, Khudoerkov R, Illarioshkin S. The Influence of Striatal Astrocyte Dysfunction on Locomotor Activity in Dopamine-depleted Rats. Basic Clin Neurosci 2021; 12:767-776. [PMID: 35693141 PMCID: PMC9168813 DOI: 10.32598/bcn.2021.1923.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/23/2019] [Accepted: 06/24/2020] [Indexed: 11/22/2022] Open
Abstract
Introduction Astrocyte dysfunction is the common pathology failing astrocyte-neuron interaction in neurological diseases, including Parkinson's Disease (PD). The present study aimed to evaluate the impacts of astrocytic dysfunction caused by striatal injections of selective glial toxin L-Aminoadipic Acid (L-AA) on the rats' locomotor activity in normal conditions and under alpha-methyl-p-tyrosine depletion of catecholamines synthesis. Methods Thirty-three male Wistar rats were used in the experiments. Intrastriatal L-AA injections (100 μg) were performed into the right striatum. Alpha-methyl-p-tyrosine (a-MT, 100 mg/kg, inhibitor of tyrosine hydroxylase) was intraperitoneally injected for catecholamine depletion. The animals were divided into 5 groups, as follows: 1. L-AA treated (n=7), 2. L-AA+a-MT treated (n=5), 3. Sham-operated (n=7), 4. Sham+a-MT treated (n=5), 5. Intact control (n=9). For assessing motor function, open field and beam walking tests were used on the third day after the operation. Neuronal and astrocyte markers (glial fibrillary acidic protein, glutamine synthetase, tyrosine hydroxylase, & neuronal nuclear antigen) were examined in the striatum by immunohistochemistry. Results Administrating L-AA led to astrocytic degeneration in the striatum. No neuronal death and disruption of dopaminergic terminals were detected. L-AA and a-MT-treated animals' distance traveled was significantly (P=0.047) shorter than the Sham-operated group injected with a-MT. In the walking beam test, the number of unilateral paw slippings was significantly (P<0.01) higher in the L-AA-treated group than Sham-operated animals. Administrating a-MT alone and L-AA did not change rats' performance in walking beam tests. Conclusion Astrocyte ablation in dopamine depleted striatum resulted in reduced motor activity and asymmetrical gait disturbances. These findings demonstrated the role of astroglia in motor function regulation in the nigrostriatal system and suggest the possible association of glial dysfunction with motor dysfunction in PD. Highlights The local administration of gliotoxin L-aminoadipate in the striatum of rats causes astrocytic degeneration without affecting the neurons and nigrostriatal fibers.The failure of astrocyte-neuron coupling in the striatum leads to motor dysfunction such as gait disturbances and bradykinesia.The influence of astrocytic degeneration on behavior is preserved and enhanced in dopamine-depleted rats. Plain Language Summary Astrocytes are the nervous system's cells supporting the function of neurons. The failure of astrocyte-neuron interaction is observed in neurological diseases, including Parkinson's disease. We induced the aminoadipate-induced rat model of astrocytic dysfunction to evaluate the role of these cells in movement regulation. In our study, astrocytic dysfunction led to gait disturbances and impaired motor function. The results suggest a possible role of glial pathology in motor impairment in parkinsonism.
Collapse
|
14
|
Zhou X, Chen Y, Peng J, Zuo M, Sun Y. Deafening-induced rapid changes to spine synaptic connectivity in the adult avian vocal basal ganglia. Integr Zool 2021; 17:1136-1146. [PMID: 34599554 DOI: 10.1111/1749-4877.12593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The basal ganglia have been implicated in auditory-dependent vocal learning and plasticity in human and songbirds, but the underlying neural phenotype remains to be clarified. Here, using confocal imaging and three-dimensional electron microscopy, we investigated striatal structural plasticity in response to hearing loss in Area X, the avian vocal basal ganglia, in adult male zebra finch (Taeniopygia guttata). We observed a rapid elongation of dendritic spines, by approximately 13%, by day 3 after deafening, and a considerable increase in spine synapse density, by approximately 61%, by day 14 after deafening, compared with the controls with an intact cochlea. These findings reveal structural sensitivity of Area X to auditory deprivation and suggest that this striatal plasticity might contribute to deafening-induced changes to learned vocal behavior.
Collapse
Affiliation(s)
- Xiaojuan Zhou
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Bejiing Normal University, Beijing, China.,Chinese Institute for Brain Research (CIBR), Beijing, China
| | - Yalan Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Bejiing Normal University, Beijing, China.,Technology Center for Protein Sciences, Tsinghua University, Beijing, China
| | - Jikan Peng
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Bejiing Normal University, Beijing, China.,School of Life Sciences, Westlake University, Hangzhou, China
| | - Mingxue Zuo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Bejiing Normal University, Beijing, China
| | - Yingyu Sun
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Bejiing Normal University, Beijing, China
| |
Collapse
|
15
|
Villalba RM, Behnke JA, Pare JF, Smith Y. Comparative Ultrastructural Analysis of Thalamocortical Innervation of the Primary Motor Cortex and Supplementary Motor Area in Control and MPTP-Treated Parkinsonian Monkeys. Cereb Cortex 2021; 31:3408-3425. [PMID: 33676368 PMCID: PMC8599722 DOI: 10.1093/cercor/bhab020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
The synaptic organization of thalamic inputs to motor cortices remains poorly understood in primates. Thus, we compared the regional and synaptic connections of vGluT2-positive thalamocortical glutamatergic terminals in the supplementary motor area (SMA) and the primary motor cortex (M1) between control and MPTP-treated parkinsonian monkeys. In controls, vGluT2-containing fibers and terminal-like profiles invaded layer II-III and Vb of M1 and SMA. A significant reduction of vGluT2 labeling was found in layer Vb, but not in layer II-III, of parkinsonian animals, suggesting a potential thalamic denervation of deep cortical layers in parkinsonism. There was a significant difference in the pattern of synaptic connectivity in layers II-III, but not in layer Vb, between M1 and SMA of control monkeys. However, this difference was abolished in parkinsonian animals. No major difference was found in the proportion of perforated versus macular post-synaptic densities at thalamocortical synapses between control and parkinsonian monkeys in both cortical regions, except for a slight increase in the prevalence of perforated axo-dendritic synapses in the SMA of parkinsonian monkeys. Our findings suggest that disruption of the thalamic innervation of M1 and SMA may underlie pathophysiological changes of the motor thalamocortical loop in the state of parkinsonism.
Collapse
Affiliation(s)
- Rosa M Villalba
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Joseph A Behnke
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Jean-Francois Pare
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
16
|
Kim E, Jeon S, An HK, Kianpour M, Yu SW, Kim JY, Rah JC, Choi H. A magnetically actuated microrobot for targeted neural cell delivery and selective connection of neural networks. SCIENCE ADVANCES 2020; 6:eabb5696. [PMID: 32978164 PMCID: PMC7518876 DOI: 10.1126/sciadv.abb5696] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/10/2020] [Indexed: 05/02/2023]
Abstract
There has been a great deal of interest in the development of technologies for actively manipulating neural networks in vitro, providing natural but simplified environments in a highly reproducible manner in which to study brain function and related diseases. Platforms for these in vitro neural networks require precise and selective neural connections at the target location, with minimal external influences, and measurement of neural activity to determine how neurons communicate. Here, we report a neuron-loaded microrobot for selective connection of neural networks via precise delivery to a gap between two neural clusters by an external magnetic field. In addition, the extracellular action potential was propagated from one cluster to the other through the neurons on the microrobot. The proposed technique shows the potential for use in experiments to understand how neurons communicate in the neural network by actively connecting neural clusters.
Collapse
Affiliation(s)
- Eunhee Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea
- DGIST-ETH Microrobot Research Center, DGIST, Daegu 42988, South Korea
| | - Sungwoong Jeon
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea
- DGIST-ETH Microrobot Research Center, DGIST, Daegu 42988, South Korea
| | - Hyun-Kyu An
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
| | | | - Seong-Woon Yu
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
| | - Jin-Young Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea.
- DGIST-ETH Microrobot Research Center, DGIST, Daegu 42988, South Korea
| | - Jong-Cheol Rah
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
- Korea Brain Research Institute, Daegu 42988, South Korea
| | - Hongsoo Choi
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea.
- DGIST-ETH Microrobot Research Center, DGIST, Daegu 42988, South Korea
- Robotics Research Center, DGIST, Daegu 42988, South Korea
| |
Collapse
|
17
|
Bensussen S, Shankar S, Ching KH, Zemel D, Ta TL, Mount RA, Shroff SN, Gritton HJ, Fabris P, Vanbenschoten H, Beck C, Man HY, Han X. A Viral Toolbox of Genetically Encoded Fluorescent Synaptic Tags. iScience 2020; 23:101330. [PMID: 32674057 PMCID: PMC7363701 DOI: 10.1016/j.isci.2020.101330] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 04/18/2020] [Accepted: 06/26/2020] [Indexed: 01/16/2023] Open
Abstract
Fibronectin intrabodies generated with mRNA display (FingRs) are a recently developed tool for labeling excitatory or inhibitory synapses, with the benefit of not altering endogenous synaptic protein expression levels or synaptic transmission. Here, we generated a viral vector FingR toolbox that allows for multi-color, neuron-type-specific labeling of excitatory or inhibitory synapses in multiple brain regions. We screened various fluorophores, FingR fusion configurations, and transcriptional control regulations in adeno-associated virus (AAV) and retrovirus vector designs. We report the development of a red FingR variant and demonstrated dual labeling of excitatory and inhibitory synapses in the same cells. Furthermore, we developed cre-inducible FingR AAV variants and demonstrated their utility, finding that the density of inhibitory synapses in aspiny striatal cholinergic interneurons remained unchanged in response to dopamine depletion. Finally, we generated FingR retroviral vectors, which enabled us to track the development of excitatory and inhibitory synapses in hippocampal adult-born granule cells.
Collapse
Affiliation(s)
- Seth Bensussen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sneha Shankar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kimberley H Ching
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Dana Zemel
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Tina L Ta
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Rebecca A Mount
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sanaya N Shroff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Howard J Gritton
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Pierre Fabris
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | | | - Connor Beck
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Schechter M, Grigoletto J, Abd-Elhadi S, Glickstein H, Friedman A, Serrano GE, Beach TG, Sharon R. A role for α-Synuclein in axon growth and its implications in corticostriatal glutamatergic plasticity in Parkinson's disease. Mol Neurodegener 2020; 15:24. [PMID: 32228705 PMCID: PMC7104492 DOI: 10.1186/s13024-020-00370-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND α-Synuclein (α-Syn) is a protein implicated in the pathogenesis of Parkinson's disease (PD). α-Syn has been shown to associate with membranes and bind acidic phospholipids. However, the physiological importance of these associations to the integrity of axons is not fully clear. METHODS Biochemical, immunohistochemical and ultrastructural analyses in cultured neurons, transgenic mouse brains, PD and control human brains. RESULTS We analyzed the ultrastructure of cross-sectioned axons localized to white matter tracts (WMTs), within the dorsal striatum of old and symptomatic α-Syn transgenic mouse brains. The analysis indicated a higher density of axons of thinner diameter. Our findings in cultured cortical neurons indicate a role for α-Syn in elongation of the main axon and its collaterals, resulting in enhanced axonal arborization. We show that α-Syn effect to enhance axonal outgrowth is mediated through its activity to regulate membrane levels of the acidic phosphatidylinositol 4,5-bisphosphate (PI4,5P2). Moreover, our findings link α-Syn- enhanced axonal growth with evidence for axonal injury. In relevance to disease mechanisms, we detect in human brains evidence for a higher degree of corticostriatal glutamatergic plasticity within WMTs at early stages of PD. However, at later PD stages, the respective WMTs in the caudate are degenerated with accumulation of Lewy pathology. CONCLUSIONS Our results show that through regulating PI4,5P2 levels, α-Syn acts to elongate the main axon and collaterals, resulting in a higher density of axons in the striatal WMTs. Based on these results we suggest a role for α-Syn in compensating mechanisms, involving corticostriatal glutamatergic plasticity, taking place early in PD.
Collapse
Affiliation(s)
- Meir Schechter
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Jessica Grigoletto
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Suaad Abd-Elhadi
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Hava Glickstein
- Electron Microscopy Unit, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Alexander Friedman
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | | | | | - Ronit Sharon
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| |
Collapse
|
19
|
Diniz LP, Matias I, Araujo APB, Garcia MN, Barros-Aragão FGQ, Alves-Leon SV, de Souza JM, Foguel D, Figueiredo CP, Braga C, Romão L, Gomes FCA. α-synuclein oligomers enhance astrocyte-induced synapse formation through TGF-β1 signaling in a Parkinson's disease model. J Neurochem 2020; 150:138-157. [PMID: 31009074 DOI: 10.1111/jnc.14710] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is characterized by selective death of dopaminergic neurons in the substantia nigra, degeneration of the nigrostriatal pathway, increases in glutamatergic synapses in the striatum and aggregation of α-synuclein. Evidence suggests that oligomeric species of α-synuclein (αSO) are the genuine neurotoxins of PD. Although several studies have supported the direct neurotoxic effects of αSO on neurons, their effects on astrocytes have not been directly addressed. Astrocytes are essential to several steps of synapse formation and function, including secretion of synaptogenic factors, control of synaptic elimination and stabilization, secretion of neural/glial modulators, and modulation of extracellular ions, and neurotransmitter levels in the synaptic cleft. Here, we show that αSO induced the astrocyte reactivity and enhanced the synaptogenic capacity of human and murine astrocytes by increasing the levels of the known synaptogenic molecule transforming growth factor beta 1 (TGF-β1). Moreover, intracerebroventricular injection of αSO in mice increased the number of astrocytes, the density of excitatory synapses, and the levels of TGF-β1 in the striatum of injected animals. Inhibition of TGF-β1 signaling impaired the effect of the astrocyte-conditioned medium on glutamatergic synapse formation in vitro and on striatal synapse formation in vivo, whereas addition of TGF-β1 protected mesencephalic neurons against synapse loss triggered by αSO. Together, our data suggest that αSO have important effects on astrocytic functions and describe TGF-β1 as a new endogenous astrocyte-derived molecule involved in the increase in striatal glutamatergic synaptic density present in early stages of PD. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14514.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Isadora Matias
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ana Paula Bérgamo Araujo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Matheus Nunes Garcia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Soniza Vieira Alves-Leon
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Jorge Marcondes de Souza
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Débora Foguel
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Carolina Braga
- Núcleo Multidisciplinar de Pesquisa em Biologia - NUMPEX-BIO, Campus Duque de Caxias Professor Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Luciana Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | |
Collapse
|
20
|
Kim AY, Oh C, Im HJ, Baek HM. Enhanced Bidirectional Connectivity of the Subthalamo-pallidal Pathway in 6-OHDA-mouse Model of Parkinson's Disease Revealed by Probabilistic Tractography of Diffusion-weighted MRI at 9.4T. Exp Neurobiol 2020; 29:80-92. [PMID: 32122110 PMCID: PMC7075660 DOI: 10.5607/en.2020.29.1.80] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/15/2020] [Accepted: 02/15/2020] [Indexed: 12/12/2022] Open
Abstract
An important challenge in Parkinson’s disease (PD) based neuroscience and neuroimaging is mapping the neuronal connectivity of the basal ganglia to understand how the disease affects brain circuitry. However, a majority of diffusion tractography studies have shown difficulties in revealing connections between distant anatomic brain regions and visualizing basal ganglia connectome. In this current study, we investigated the differences in basal ganglia connectivity between 6-OHDA induced ex-vivo PD mouse model and normal ex-vivo mouse model by using diffusion tensor imaging tractography from diffusion-weighted images obtained with a high resolution 9.4 T MR scanner. Connectivity pattern of the basal ganglia were compared between five 6-OHDA and five control ex-vivo mouse brains using results of probabilistic tractography generated with PROBTRACKX. When compared with control mouse, 6-OHDA mouse showed significant enhancements to motor territory-related subthalamo-pallidal and pallido-subthalamic connectivity. Multi-fiber tractography combined with diffusion MRI data has the potential to help recognize the abnormalities found in connectivity of psychiatric and neurologic disease models.
Collapse
Affiliation(s)
- A-Yoon Kim
- Department of Health Science and Technology, GAIHST, Gachon University, Incheon 21936, Korea
| | - Chiwoo Oh
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 16229, Korea
| | - Hyung-Jun Im
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 16229, Korea
| | - Hyeon-Man Baek
- Department of Health Science and Technology, GAIHST, Gachon University, Incheon 21936, Korea.,Lee Gil Ya Cancer & Diabetes Institute, Gachon University, Incheon 21999, Korea
| |
Collapse
|
21
|
Lee S, Negishi M, Urakubo H, Kasai H, Ishii S. Mu-net: Multi-scale U-net for two-photon microscopy image denoising and restoration. Neural Netw 2020; 125:92-103. [PMID: 32078964 DOI: 10.1016/j.neunet.2020.01.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 12/18/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Advances in two two-photon microscopy (2PM) have made three-dimensional (3D) neural imaging of deep cortical regions possible. However, 2PM often suffers from poor image quality because of various noise factors, including blur, white noise, and photo bleaching. In addition, the effectiveness of the existing image processing methods is limited because of the special features of 2PM images such as deeper tissue penetration but higher image noises owing to rapid laser scanning. To address the denoising problems in 2PM 3D images, we present a new algorithm based on deep convolutional neural networks (CNNs). The proposed model consists of multiple U-nets in which an individual U-net removes noises at different scales and then yields a performance improvement based on a coarse-to-fine strategy. Moreover, the constituent CNNs employ fully 3D convolution operations. Such an architecture enables the proposed model to facilitate end-to-end learning without any pre/post processing. Based on the experiments on 2PM image denoising, we observed that our new algorithm demonstrates substantial performance improvements over other baseline methods.
Collapse
Affiliation(s)
- Sehyung Lee
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Japan.
| | - Makiko Negishi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Japan
| | - Hidetoshi Urakubo
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Japan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Japan
| | - Shin Ishii
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Japan; Advanced Telecommunications Research Institute International (ATR), Japan
| |
Collapse
|
22
|
Modified Glutamatergic Postsynapse in Neurodegenerative Disorders. Neuroscience 2019; 454:116-139. [PMID: 31887357 DOI: 10.1016/j.neuroscience.2019.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/02/2019] [Accepted: 12/02/2019] [Indexed: 01/27/2023]
Abstract
The postsynaptic density (PSD) is a complex subcellular domain important for postsynaptic signaling, function, and plasticity. The PSD is present at excitatory synapses and specialized to allow for precise neuron-to-neuron transmission of information. The PSD is localized immediately underneath the postsynaptic membrane forming a major protein network that regulates postsynaptic signaling and synaptic plasticity. Glutamatergic synaptic dysfunction affecting PSD morphology and signaling events have been described in many neurodegenerative disorders, either sporadic or familial forms. Thus, in this review we describe the main protein players forming the PSD and their activity, as well as relevant modifications in key components of the postsynaptic architecture occurring in Huntington's, Parkinson's and Alzheimer's diseases.
Collapse
|
23
|
Swain AJ, Galvan A, Wichmann T, Smith Y. Structural plasticity of GABAergic and glutamatergic networks in the motor thalamus of parkinsonian monkeys. J Comp Neurol 2019; 528:1436-1456. [PMID: 31808567 DOI: 10.1002/cne.24834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/10/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022]
Abstract
In the primate thalamus, the parvocellular ventral anterior nucleus (VApc) and the centromedian nucleus (CM) receive GABAergic projections from the internal globus pallidus (GPi) and glutamatergic inputs from motor cortices. In this study, we used electron microscopy to assess potential structural changes in GABAergic and glutamatergic microcircuits in the VApc and CM of MPTP-treated parkinsonian monkeys. The intensity of immunostaining for GABAergic markers in VApc and CM did not differ between control and parkinsonian monkeys. In the electron microscope, three major types of terminals were identified in both nuclei: (a) vesicular glutamate transporter 1 (vGluT1)-positive terminals forming asymmetric synapses (type As), which originate from the cerebral cortex, (b) GABAergic terminals forming single symmetric synapses (type S1), which likely arise from the reticular nucleus and GABAergic interneurons, and (c) GABAergic terminals forming multiple symmetric synapses (type S2), which originate from GPi. The density of As terminals outnumbered that of S1 and S2 terminals in VApc and CM of control and parkinsonian animals. No significant change was found in the abundance and synaptic connectivity of S1 and S2 terminals in VApc or CM of MPTP-treated monkeys, while the prevalence of "As" terminals in VApc of parkinsonian monkeys was 51.4% lower than in controls. The cross-sectional area of vGluT1-positive boutons in both VApc and CM of parkinsonian monkeys was significantly larger than in controls, but their pattern of innervation of thalamic cells was not altered. Our findings suggest that the corticothalamic system undergoes significant synaptic remodeling in the parkinsonian state.
Collapse
Affiliation(s)
- Ashley J Swain
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Atlanta, Georgia.,Udall Center of Excellence for Parkinson's Disease Research, Atlanta, Georgia
| | - Adriana Galvan
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Atlanta, Georgia.,Udall Center of Excellence for Parkinson's Disease Research, Atlanta, Georgia.,Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| | - Thomas Wichmann
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Atlanta, Georgia.,Udall Center of Excellence for Parkinson's Disease Research, Atlanta, Georgia.,Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Atlanta, Georgia.,Udall Center of Excellence for Parkinson's Disease Research, Atlanta, Georgia.,Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
24
|
Villalba RM, Pare JF, Lee S, Lee S, Smith Y. Thalamic degeneration in MPTP-treated Parkinsonian monkeys: impact upon glutamatergic innervation of striatal cholinergic interneurons. Brain Struct Funct 2019; 224:3321-3338. [PMID: 31679085 PMCID: PMC6878768 DOI: 10.1007/s00429-019-01967-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
In both Parkinson's disease (PD) patients and MPTP-treated non-human primates, there is a profound neuronal degeneration of the intralaminar centromedian/parafascicular (CM/Pf) thalamic complex. Although this thalamic pathology has long been established in PD (and other neurodegenerative disorders), the impact of CM/Pf cell loss on the integrity of the thalamo-striatal glutamatergic system and its regulatory functions upon striatal neurons remain unknown. In the striatum, cholinergic interneurons (ChIs) are important constituents of the striatal microcircuitry and represent one of the main targets of CM/Pf-striatal projections. Using light and electron microscopy approaches, we have analyzed the potential impact of CM/Pf neuronal loss on the anatomy of the synaptic connections between thalamic terminals (vGluT2-positive) and ChIs neurons in the striatum of parkinsonian monkeys treated chronically with MPTP. The following conclusions can be drawn from our observations: (1) as reported in PD patients, and in our previous monkey study, CM/Pf neurons undergo profound degeneration in monkeys chronically treated with low doses of MPTP. (2) In the caudate (head and body) nucleus of parkinsonian monkeys, there is an increased density of ChIs. (3) Despite the robust loss of CM/Pf neurons, no significant change was found in the density of thalamostriatal (vGluT2-positive) terminals, and in the prevalence of vGluT2-positive terminals in contact with ChIs in parkinsonian monkeys. These findings provide new information about the state of thalamic innervation of the striatum in parkinsonian monkeys with CM/Pf degeneration, and bring up an additional level of intricacy to the consequences of thalamic pathology upon the functional microcircuitry of the thalamostriatal system in parkinsonism. Future studies are needed to assess the importance of CM/Pf neuronal loss, and its potential consequences on the neuroplastic changes induced in the synaptic organization of the thalamostriatal system, in the development of early cognitive impairments in PD.
Collapse
Affiliation(s)
- Rosa M Villalba
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 303, USA.
- UDALL Center for Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.
| | - Jean-Francois Pare
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 303, USA
- UDALL Center for Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA
| | - Solah Lee
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 303, USA
- UDALL Center for Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA
| | - Sol Lee
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 303, USA
- UDALL Center for Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 303, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
- UDALL Center for Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA
| |
Collapse
|
25
|
Sikora J, Kieffer BL, Paoletti P, Ouagazzal AM. Synaptic zinc contributes to motor and cognitive deficits in 6-hydroxydopamine mouse models of Parkinson's disease. Neurobiol Dis 2019; 134:104681. [PMID: 31759136 DOI: 10.1016/j.nbd.2019.104681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/05/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022] Open
Abstract
Hyperactivity of glutamatergic corticostrial pathways is recognized as a key pathophysiological mechanism contributing to development of PD symptoms and dopaminergic neurotoxicity. Subset of corticostriatal projection neurons uses Zn2+ as a co-transmitter alongside glutamate, but the role of synaptically released Zn2+ in PD remains unexplored. We used genetically modified mice and pharmacological tools in combination with 6-hydroxydopamine (6-OHDA) lesion models of PD to investigate the contribution of synaptic zinc to disease associated behavioral deficits and neurodegeneration. Vesicular zinc transporter-3 (ZnT3) knockout mice lacking releasable Zn2+ were more resistant to locomotor deficit and memory impairment of nigrostriatal dopamine (DA) denervation compared to wildtype littermates. The loss of striatal dopaminergic fibers was comparable between genotypes, indicating that synaptically released Zn2+ contributes to behavioral deficits but not neurotoxic effects of 6-OHDA. To gain further insight into the mechanisms of Zn2+ actions, we used the extracellular Zn2+ chelator CaEDTA and knock-in mice lacking the high affinity Zn2+ inhibition of GluN2A-containing NMDA receptors (GluN2A-NMDARs). Acute chelation of extracellular Zn2+ in the striatum restored locomotor deficit of 6-OHDA lesion, confirming that synaptic Zn2+ suppresses locomotor behavior. Disruption of the Zn2+-GluN2A interaction had, on the other hand, no impact on locomotor deficit or neurotoxic effect of 6-OHDA. Collectively, these findings provide clear evidence for the implication of striatal synaptic Zn2+ in the pathophysiology of PD. They unveil that synaptic Zn2+ plays predominantly a detrimental role by promoting motor and cognitive deficits caused by nigrostriatal DA denervation, pointing towards new therapeutic interventions.
Collapse
Affiliation(s)
- Joanna Sikora
- Laboratoire de Neurosciences Cognitives, Aix-Marseille Univ, CNRS, LNC, UMR 7291, 13331 Marseille, France; Aix-marseille Université, Marseille, France
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Canada
| | - Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, Paris, France
| | - Abdel-Mouttalib Ouagazzal
- Laboratoire de Neurosciences Cognitives, Aix-Marseille Univ, CNRS, LNC, UMR 7291, 13331 Marseille, France.
| |
Collapse
|
26
|
Foffani G, Obeso JA. A Cortical Pathogenic Theory of Parkinson's Disease. Neuron 2019; 99:1116-1128. [PMID: 30236282 DOI: 10.1016/j.neuron.2018.07.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/14/2018] [Accepted: 07/17/2018] [Indexed: 01/17/2023]
Abstract
In Parkinson's disease, the progressive neurodegeneration of nigrostriatal dopaminergic neurons in the substantia nigra pars compacta (SNc) is associated with classic motor features, which typically have a focal onset. Since a defined somatotopic arrangement in the SNc has not been recognized, this focal motor onset is unexplained and hardly justified by current pathogenic theories of bottom-up disease progression (Braak's hypothesis, prionopathy). Here we propose that corticostriatal activity may represent a critical somatotopic "stressor" for nigrostriatal terminals, ultimately driving retrograde nigrostriatal degeneration and leading to focal motor onset and progression of Parkinson's disease. As a pathogenic mechanism, corticostriatal activity may promote secretion of striatal extracellular alpha-synuclein, favoring its pathological aggregation at vulnerable dopaminergic synapses. A similar pathogenic process may occur at corticofugal projections to the medulla oblongata and other vulnerable structures, thereby contributing to the bottom-up progression of Lewy pathology. This cortical pathogenesis may co-exist with bottom-up mechanisms, adding an integrative top-down perspective to the quest for the factors that impinge upon the vulnerability of dopaminergic cells in the onset and progression of Parkinson's disease.
Collapse
Affiliation(s)
- Guglielmo Foffani
- CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU-San Pablo, Madrid, Spain; Hospital Nacional de Parapléjicos, Toledo, Spain.
| | - José A Obeso
- CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU-San Pablo, Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
27
|
Wichmann T. Changing views of the pathophysiology of Parkinsonism. Mov Disord 2019; 34:1130-1143. [PMID: 31216379 DOI: 10.1002/mds.27741] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/11/2022] Open
Abstract
Studies of the pathophysiology of parkinsonism (specifically akinesia and bradykinesia) have a long history and primarily model the consequences of dopamine loss in the basal ganglia on the function of the basal ganglia/thalamocortical circuit(s). Changes of firing rates of individual nodes within these circuits were originally considered central to parkinsonism. However, this view has now given way to the belief that changes in firing patterns within the basal ganglia and related nuclei are more important, including the emergence of burst discharges, greater synchrony of firing between neighboring neurons, oscillatory activity patterns, and the excessive coupling of oscillatory activities at different frequencies. Primarily focusing on studies obtained in nonhuman primates and human patients with Parkinson's disease, this review summarizes the current state of this field and highlights several emerging areas of research, including studies of the impact of the heterogeneity of external pallidal neurons on parkinsonism, the importance of extrastriatal dopamine loss, parkinsonism-associated synaptic and morphologic plasticity, and the potential role(s) of the cerebellum and brainstem in the motor dysfunction of Parkinson's disease. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Thomas Wichmann
- Department of Neurology/School of Medicine and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Keifman E, Ruiz-DeDiego I, Pafundo DE, Paz RM, Solís O, Murer MG, Moratalla R. Optostimulation of striatonigral terminals in substantia nigra induces dyskinesia that increases after L-DOPA in a mouse model of Parkinson's disease. Br J Pharmacol 2019; 176:2146-2161. [PMID: 30895594 DOI: 10.1111/bph.14663] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/08/2019] [Accepted: 02/20/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND AND PURPOSE L-DOPA-induced dyskinesia (LID) remains a major complication of L-DOPA therapy in Parkinson's disease. LID is believed to result from inhibition of substantia nigra reticulata (SNr) neurons by GABAergic striatal projection neurons that become supersensitive to dopamine receptor stimulation after severe nigrostriatal degeneration. Here, we asked if stimulation of direct medium spiny neuron (dMSN) GABAergic terminals at the SNr can produce a full dyskinetic state similar to that induced by L-DOPA. EXPERIMENTAL APPROACH Adult C57BL6 mice were lesioned with 6-hydroxydopamine in the medial forebrain bundle. Channel rhodopsin was expressed in striatonigral terminals by ipsilateral striatal injection of adeno-associated viral particles under the CaMKII promoter. Optic fibres were implanted on the ipsilateral SNr. Optical stimulation was performed before and 24 hr after three daily doses of L-DOPA at subthreshold and suprathreshold dyskinetic doses. We also examined the combined effect of light stimulation and an acute L-DOPA challenge. KEY RESULTS Optostimulation of striatonigral terminals inhibited SNr neurons and induced all dyskinesia subtypes (optostimulation-induced dyskinesia [OID]) in 6-hydroxydopamine animals, but not in sham-lesioned animals. Additionally, chronic L-DOPA administration sensitised dyskinetic responses to striatonigral terminal optostimulation, as OIDs were more severe 24 hr after L-DOPA administration. Furthermore, L-DOPA combined with light stimulation did not result in higher dyskinesia scores than OID alone, suggesting that optostimulation has a masking effect on LID. CONCLUSION AND IMPLICATIONS This work suggests that striatonigral inhibition of basal ganglia output (SNr) is a decisive mechanism mediating LID and identifies the SNr as a target for managing LID.
Collapse
Affiliation(s)
- Ettel Keifman
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Irene Ruiz-DeDiego
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Diego Esteban Pafundo
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Rodrigo Manuel Paz
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| |
Collapse
|
29
|
Pelassa S, Guidolin D, Venturini A, Averna M, Frumento G, Campanini L, Bernardi R, Cortelli P, Buonaura GC, Maura G, Agnati LF, Cervetto C, Marcoli M. A2A-D2 Heteromers on Striatal Astrocytes: Biochemical and Biophysical Evidence. Int J Mol Sci 2019; 20:ijms20102457. [PMID: 31109007 PMCID: PMC6566402 DOI: 10.3390/ijms20102457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Our previous findings indicate that A2A and D2 receptors are co-expressed on adult rat striatal astrocytes and on the astrocyte processes, and that A2A-D2 receptor–receptor interaction can control the release of glutamate from the processes. Functional evidence suggests that the receptor–receptor interaction was based on heteromerization of native A2A and D2 receptors at the plasma membrane of striatal astrocyte processes. We here provide biochemical and biophysical evidence confirming that receptor–receptor interaction between A2A and D2 receptors at the astrocyte plasma membrane is based on A2A-D2 heteromerization. To our knowledge, this is the first direct demonstration of the ability of native A2A and D2 receptors to heteromerize on glial cells. As striatal astrocytes are recognized to be involved in Parkinson’s pathophysiology, the findings that adenosine A2A and dopamine D2 receptors can form A2A-D2 heteromers on the astrocytes in the striatum (and that these heteromers can play roles in the control of the striatal glutamatergic transmission) may shed light on the molecular mechanisms involved in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Simone Pelassa
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Via Gabelli 63, 35122 Padova, Italy.
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Monica Averna
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy.
| | - Giulia Frumento
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Letizia Campanini
- Division of Experimental Oncology, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy.
| | - Rosa Bernardi
- Division of Experimental Oncology, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy.
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum-University of Bologna, Via Altura 3, 40139 Bologna, Italy.
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, 40139 Bologna, Italy.
| | - Giovanna Calandra Buonaura
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum-University of Bologna, Via Altura 3, 40139 Bologna, Italy.
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, 40139 Bologna, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy.
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 171 65 Stockholm, Sweden.
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
- Centre of Excellence for Biomedical Research CEBR, University of Genova, Viale Benedetto XV, 5, 16132 Genova, Italy.
| |
Collapse
|
30
|
Shim JH, Im SJ, Kim AY, Kim YT, Kim EB, Baek HM. Generation of Mouse Basal Ganglia Diffusion Tractography Using 9.4T MRI. Exp Neurobiol 2019; 28:300-310. [PMID: 31138997 PMCID: PMC6526107 DOI: 10.5607/en.2019.28.2.300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/05/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Over the years, diffusion tractography has seen increasing use for comparing minute differences in connectivity of brain structures in neurodegenerative diseases and treatments. Studies on connectivity between basal ganglia has been a focal point for studying the effects of diseases such as Parkinson's and Alzheimer's, as well as the effects of treatments such as deep brain stimulation. Additionally, in previous studies, diffusion tractography was utilized in disease mouse models to identify white matter alterations, as well as biomarkers that occur in the progression of disease. However, despite the extensive use of mouse models to study model diseases, the structural connectivity of the mouse basal ganglia has been inadequately explored. In this study, we present the methodology of segmenting the basal ganglia of a mouse brain, then generating diffusion tractography between the segmented basal ganglia structures. Additionally, we compare the relative levels of connectivity of connecting fibers between each basal ganglia structure, as well as visualize the shapes of each connection. We believe that our results and future studies utilizing diffusion tractography will be beneficial for properly assessing some of the connectivity changes that are found in the basal ganglia of various mouse models.
Collapse
Affiliation(s)
- Jae-Hyuk Shim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Sang-Jin Im
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - A-Yoon Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Yong-Tae Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Eun Bee Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Hyeon-Man Baek
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| |
Collapse
|
31
|
Zheng X, Huang Z, Zhu Y, Liu B, Chen Z, Chen T, Jia L, Li Y, Lei W. Increase in Glutamatergic Terminals in the Striatum Following Dopamine Depletion in a Rat Model of Parkinson's Disease. Neurochem Res 2019; 44:1079-1089. [PMID: 30715657 DOI: 10.1007/s11064-019-02739-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022]
Abstract
Dopaminergic neuron degeneration is known to give rise to dendrite injury and spine loss of striatal neurons, however, changes of intrastriatal glutamatergic terminals and their synapses after 6-hydroxydopamine (6OHDA)-induced dopamine (DA)-depletion remains controversial. To confirm the effect of striatal DA-depletion on the morphology and protein levels of corticostriatal and thalamostriatal glutamatergic terminals and synapses, immunohistochemistry, immuno-electron microscope (EM), western blotting techniques were performed on Parkinson's disease rat models in this study. The experimental results of this study showed that: (1) 6OHDA-induced DA-depletion resulted in a remarkable increase of Vesicular glutamate transporter 1 (VGlut1) + and Vesicular glutamate transporter 2 (VGlut2)+ terminal densities at both the light microscope (LM) and EM levels, and VGlut1+ and VGlut2+ terminal sizes were shown to be enlarged by immuno-EM; (2) Striatal DA-depletion resulted in a decrease in both the total and axospinous terminal fractions of VGlut1+ terminals, but the axodendritic terminal fraction was not significantly different from the control group. However, total, axospinous and axodendritic terminal fractions for VGlut2+ terminals declined significantly after striatal DA-depletion. (3) Western blotting data showed that striatal DA-depletion up-regulated the expression levels of the VGlut1 and VGlut2 proteins. These results suggest that 6OHDA-induced DA-depletion affects corticostriatal and thalamostriatal glutamatergic synaptic inputs, which are involved in the pathological process of striatal neuron injury induced by DA-depletion.
Collapse
Affiliation(s)
- Xuefeng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaofeng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Medicine, College of Medicine, Jishou University, Jishou, China
| | - Bingbing Liu
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yanmei Li
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
32
|
Cervetto C, Venturini A, Guidolin D, Maura G, Passalacqua M, Tacchetti C, Cortelli P, Genedani S, Candiani S, Ramoino P, Pelassa S, Marcoli M, Agnati LF. Homocysteine and A2A-D2 Receptor-Receptor Interaction at Striatal Astrocyte Processes. J Mol Neurosci 2018; 65:456-466. [PMID: 30030763 DOI: 10.1007/s12031-018-1120-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/11/2018] [Indexed: 01/03/2023]
Abstract
The interaction between adenosine A2A and dopamine D2 receptors in striatal neurons is a well-established phenomenon and has opened up new perspectives on the molecular mechanisms involved in Parkinson's disease. However, it has barely been investigated in astrocytes. Here, we show by immunofluorescence that both A2A and D2 receptors are expressed in adult rat striatal astrocytes in situ, and investigate on presence, function, and interactions of the receptors in the astrocyte processes-acutely prepared from the adult rat striatum-and on the effects of homocysteine on the A2A-D2 receptor-receptor interaction. We found that A2A and D2 receptors were co-expressed on vesicular glutamate transporter-1-positive astrocyte processes, and confirmed that A2A-D2 receptor-receptor interaction controlled glutamate release-assessed by measuring the [3H]D-aspartate release-from the processes. The complexity of A2A-D2 receptor-receptor interaction is suggested by the effect of intracellular homocysteine, which reduced D2-mediated inhibition of glutamate release (homocysteine allosteric action on D2), without interfering with the A2A-mediated antagonism of the D2 effect (maintained A2A-D2 interaction). Our findings indicate the crucial integrative role of A2A-D2 molecular circuits at the plasma membrane of striatal astrocyte processes. The fact that homocysteine reduced D2-mediated inhibition of glutamate release could provide new insights into striatal astrocyte-neuron intercellular communications. As striatal astrocytes are recognized to be involved in Parkinson's pathophysiology, these findings may shed light on the pathogenic mechanisms of the disease and contribute to the development of new drugs for its treatment.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Padua, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, Section of Biochemistry, and Italian Institute of Biostructures and Biosystems, University of Genova, Genoa, Italy
| | - Carlo Tacchetti
- Experimental Imaging Center, Scientific Institute San Raffaele, Milan, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Susanna Genedani
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Paola Ramoino
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Simone Pelassa
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy. .,Centre of Excellence for Biomedical Research CEBR, University of Genova, Genoa, Italy.
| | - Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Bhattacharya S, Ma Y, Dunn AR, Bradner JM, Scimemi A, Miller GW, Traynelis SF, Wichmann T. NMDA receptor blockade ameliorates abnormalities of spike firing of subthalamic nucleus neurons in a parkinsonian nonhuman primate. J Neurosci Res 2018; 96:1324-1335. [PMID: 29577359 PMCID: PMC5980712 DOI: 10.1002/jnr.24230] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/25/2018] [Accepted: 02/08/2018] [Indexed: 12/21/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ion channels comprising tetrameric assemblies of GluN1 and GluN2 receptor subunits that mediate excitatory neurotransmission in the central nervous system. Of the four different GluN2 subunits, the GluN2D subunit-containing NMDARs have been suggested as a target for antiparkinsonian therapy because of their expression pattern in some of the basal ganglia nuclei that show abnormal firing patterns in the parkinsonian state, specifically the subthalamic nucleus (STN). In this study, we demonstrate that blockade of NMDARs altered spike firing in the STN in a male nonhuman primate that had been rendered parkinsonian by treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. In accompanying experiments in male rodents, we found that GluN2D-NMDAR expression in the STN was reduced in acutely or chronically dopamine-depleted animals. Taken together, our data suggest that blockade of NMDARs in the STN may be a viable antiparkinsonian strategy, but that the ultimate success of this approach may be complicated by parkinsonism-associated changes in NMDAR expression in the STN.
Collapse
Affiliation(s)
| | - Yuxian Ma
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Amy R Dunn
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Joshua M Bradner
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Annalisa Scimemi
- Department of Biology, State University of New York at Albany, Albany, New York
| | - Gary W Miller
- Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
- Morris K. Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| |
Collapse
|
34
|
Starr A, Sattler R. Synaptic dysfunction and altered excitability in C9ORF72 ALS/FTD. Brain Res 2018; 1693:98-108. [PMID: 29453960 DOI: 10.1016/j.brainres.2018.02.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 02/08/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by a progressive degeneration of upper and lower motor neurons, resulting in fatal paralysis due to denervation of the muscle. Due to genetic, pathological and symptomatic overlap, ALS is now considered a spectrum disease together with frontotemporal dementia (FTD), the second most common cause of dementia in individuals under the age of 65. Interestingly, in both diseases, there is a large prevalence of RNA binding proteins (RBPs) that are mutated and considered disease-causing, or whose dysfunction contribute to disease pathogenesis. The most common shared genetic mutation in ALS/FTD is a hexanucleuotide repeat expansion within intron 1 of C9ORF72 (C9). Three potentially overlapping, putative toxic mechanisms have been proposed: loss of function due to haploinsufficient expression of the C9ORF72 mRNA, gain of function of the repeat RNA aggregates, or RNA foci, and repeat-associated non-ATG-initiated translation (RAN) of the repeat RNA into toxic dipeptide repeats (DPRs). Regardless of the causative mechanism, disease symptoms are ultimately caused by a failure of neurotransmission in three regions: the brain, the spinal cord, and the neuromuscular junction. Here, we review C9 ALS/FTD-associated synaptic dysfunction and aberrant neuronal excitability in these three key regions, focusing on changes in morphology and synapse formation, excitability, and excitotoxicity in patients, animal models, and in vitro models. We compare these deficits to those seen in other forms of ALS and FTD in search of shared pathways, and discuss the potential targeting of synaptic dysfunctions for therapeutic intervention in ALS and FTD patients.
Collapse
Affiliation(s)
- Alexander Starr
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States
| | - Rita Sattler
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States.
| |
Collapse
|
35
|
Hou L, Chen W, Liu X, Qiao D, Zhou FM. Exercise-Induced Neuroprotection of the Nigrostriatal Dopamine System in Parkinson's Disease. Front Aging Neurosci 2017; 9:358. [PMID: 29163139 PMCID: PMC5675869 DOI: 10.3389/fnagi.2017.00358] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/19/2017] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies indicate that physical activity and exercise may reduce the risk of developing Parkinson's disease (PD), and clinical observations suggest that physical exercise can reduce the motor symptoms in PD patients. In experimental animals, a profound observation is that exercise of appropriate timing, duration, and intensity can reduce toxin-induced lesion of the nigrostriatal dopamine (DA) system in animal PD models, although negative results have also been reported, potentially due to inappropriate timing and intensity of the exercise regimen. Exercise may also minimize DA denervation-induced medium spiny neuron (MSN) dendritic atrophy and other abnormalities such as enlarged corticostriatal synapse and abnormal MSN excitability and spiking activity. Taken together, epidemiological studies, clinical observations, and animal research indicate that appropriately dosed physical activity and exercise may not only reduce the risk of developing PD in vulnerable populations but also benefit PD patients by potentially protecting the residual DA neurons or directly restoring the dysfunctional cortico-basal ganglia motor control circuit, and these benefits may be mediated by exercise-triggered production of endogenous neuroprotective molecules such as neurotrophic factors. Thus, exercise is a universally available, side effect-free medicine that should be prescribed to vulnerable populations as a preventive measure and to PD patients as a component of treatment. Future research needs to establish standardized exercise protocols that can reliably induce DA neuron protection, enabling the delineation of the underlying cellular and molecular mechanisms that in turn can maximize exercise-induced neuroprotection and neurorestoration in animal PD models and eventually in PD patients.
Collapse
Affiliation(s)
- Lijuan Hou
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Wei Chen
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China.,Department of Exercise and Rehabilitation, Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Xiaoli Liu
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Decai Qiao
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Fu-Ming Zhou
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN, United States
| |
Collapse
|
36
|
Blesa J, Trigo-Damas I, Dileone M, Del Rey NLG, Hernandez LF, Obeso JA. Compensatory mechanisms in Parkinson's disease: Circuits adaptations and role in disease modification. Exp Neurol 2017; 298:148-161. [PMID: 28987461 DOI: 10.1016/j.expneurol.2017.10.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
The motor features of Parkinson's disease (PD) are well known to manifest only when striatal dopaminergic deficit reaches 60-70%. Thus, PD has a long pre-symptomatic and pre-motor evolution during which compensatory mechanisms take place to delay the clinical onset of disabling manifestations. Classic compensatory mechanisms have been attributed to changes and adjustments in the nigro-striatal system, such as increased neuronal activity in the substantia nigra pars compacta and enhanced dopamine synthesis and release in the striatum. However, it is not so clear currently that such changes occur early enough to account for the pre-symptomatic period. Other possible mechanisms relate to changes in basal ganglia and motor cortical circuits including the cerebellum. However, data from early PD patients are difficult to obtain as most studies have been carried out once the diagnosis and treatments have been established. Likewise, putative compensatory mechanisms taking place throughout disease evolution are nearly impossible to distinguish by themselves. Here, we review the evidence for the role of the best known and other possible compensatory mechanisms in PD. We also discuss the possibility that, although beneficial in practical terms, compensation could also play a deleterious role in disease progression.
Collapse
Affiliation(s)
- Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Inés Trigo-Damas
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Michele Dileone
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Natalia Lopez-Gonzalez Del Rey
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ledia F Hernandez
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
37
|
Bahuguna J, Tetzlaff T, Kumar A, Hellgren Kotaleski J, Morrison A. Homologous Basal Ganglia Network Models in Physiological and Parkinsonian Conditions. Front Comput Neurosci 2017; 11:79. [PMID: 28878643 PMCID: PMC5572265 DOI: 10.3389/fncom.2017.00079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/04/2017] [Indexed: 01/19/2023] Open
Abstract
The classical model of basal ganglia has been refined in recent years with discoveries of subpopulations within a nucleus and previously unknown projections. One such discovery is the presence of subpopulations of arkypallidal and prototypical neurons in external globus pallidus, which was previously considered to be a primarily homogeneous nucleus. Developing a computational model of these multiple interconnected nuclei is challenging, because the strengths of the connections are largely unknown. We therefore use a genetic algorithm to search for the unknown connectivity parameters in a firing rate model. We apply a binary cost function derived from empirical firing rate and phase relationship data for the physiological and Parkinsonian conditions. Our approach generates ensembles of over 1,000 configurations, or homologies, for each condition, with broad distributions for many of the parameter values and overlap between the two conditions. However, the resulting effective weights of connections from or to prototypical and arkypallidal neurons are consistent with the experimental data. We investigate the significance of the weight variability by manipulating the parameters individually and cumulatively, and conclude that the correlation observed between the parameters is necessary for generating the dynamics of the two conditions. We then investigate the response of the networks to a transient cortical stimulus, and demonstrate that networks classified as physiological effectively suppress activity in the internal globus pallidus, and are not susceptible to oscillations, whereas parkinsonian networks show the opposite tendency. Thus, we conclude that the rates and phase relationships observed in the globus pallidus are predictive of experimentally observed higher level dynamical features of the physiological and parkinsonian basal ganglia, and that the multiplicity of solutions generated by our method may well be indicative of a natural diversity in basal ganglia networks. We propose that our approach of generating and analyzing an ensemble of multiple solutions to an underdetermined network model provides greater confidence in its predictions than those derived from a unique solution, and that projecting such homologous networks on a lower dimensional space of sensibly chosen dynamical features gives a better chance than a purely structural analysis at understanding complex pathologies such as Parkinson's disease.
Collapse
Affiliation(s)
- Jyotika Bahuguna
- Institute of Neuroscience and Medicine (INM-6), Institute for Advanced Simulation (IAS-6), JARA Brain Institute I, Jülich Research CenterJülich, Germany
| | - Tom Tetzlaff
- Institute of Neuroscience and Medicine (INM-6), Institute for Advanced Simulation (IAS-6), JARA Brain Institute I, Jülich Research CenterJülich, Germany
| | - Arvind Kumar
- Computational Science and Technology, School of Computer Science and Communication, KTH Royal Institute of TechnologyStockholm, Sweden.,Faculty of Biology, Bernstein Center Freiburg, University of FreiburgFreiburg, Germany
| | - Jeanette Hellgren Kotaleski
- Computational Science and Technology, School of Computer Science and Communication, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Abigail Morrison
- Institute of Neuroscience and Medicine (INM-6), Institute for Advanced Simulation (IAS-6), JARA Brain Institute I, Jülich Research CenterJülich, Germany.,Faculty of Biology, Bernstein Center Freiburg, University of FreiburgFreiburg, Germany.,Institute for Cognitive Neurosciences, Ruhr UniversityBochum, Germany
| |
Collapse
|
38
|
Exercise in an animal model of Parkinson's disease: Motor recovery but not restoration of the nigrostriatal pathway. Neuroscience 2017; 359:224-247. [PMID: 28754312 DOI: 10.1016/j.neuroscience.2017.07.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 11/23/2022]
Abstract
Many clinical studies have reported on the benefits of exercise therapy in patients with Parkinson's disease (PD). Exercise cannot stop the progression of PD or facilitate the recovery of dopamine (DA) neurons in the substantia nigra pars compacta (SNpc) (Bega et al., 2014). To tease apart this paradox, we utilized a progressive MPTP (1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine) mouse model in which we initiated 4weeks of treadmill exercise after the completion of toxin administration (i.e., restoration). We found in our MPTP/exercise (MPTP+EX) group several measures of gait function that recovered compared to the MPTP only group. Although there was a small recovery of tyrosine hydroxylase (TH) positive DA neurons in the SNpc and terminals in the striatum, this increase was not statistically significant. These small changes in TH could not explain the improvement of motor function. The MPTP group had a significant 170% increase in the glycosylated/non-glycosylated dopamine transporter (DAT) and a 200% increase in microglial marker, IBA-1, in the striatum. The MPTP+EX group showed a nearly full recovery of these markers back to the vehicle levels. There was an increase in GLT-1 levels in the striatum due to exercise, with no change in striatal BDNF protein expression. Our data suggest that motor recovery was not prompted by any significant restoration of DA neurons or terminals, but rather the recovery of DAT and dampening the inflammatory response. Although exercise does not promote recovery of nigrostriatal DA, it should be used in conjunction with pharmaceutical methods for controlling PD symptoms.
Collapse
|
39
|
Wang X, Han C, Xu Y, Wu K, Chen S, Hu M, Wang L, Ye Y, Ye F. Synthesis and Evaluation of Phenylxanthine Derivatives as Potential Dual A2AR Antagonists/MAO-B Inhibitors for Parkinson's Disease. Molecules 2017. [PMID: 28629145 PMCID: PMC6152622 DOI: 10.3390/molecules22061010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The aim of this research was to prove the speculation that phenylxanthine (PX) derivatives possess adenosine A2A receptor (A2AR)-blocking properties and to screening and evaluate these PX derivatives as dual A2AR antagonists/MAO-B inhibitors for Parkinson′s disease. To explore this hypothesis, two series of PX derivatives were prepared and their antagonism against A2AR and inhibition against MAO-B were determined in vitro. In order to evaluate further the antiparkinsonian properties, pharmacokinetic and haloperidol-induced catalepsy experiments were carried out in vivo. The PX-D and PX-E analogues acted as potent A2AR antagonists with Ki values ranging from 0.27 to 10 μM, and these analogues displayed relatively mild MAO-B inhibition potencies, with inhibitor dissociation constants (Ki values) ranging from 0.25 to 10 μM. Further, the compounds PX-D-P6 and PX-E-P8 displayed efficacious antiparkinsonian properties in haloperidol-induced catalepsy experiments, verifying that these two compounds were potent A2AR antagonists and MAO-B inhibitors. We conclude that PX-D and PX-E analogues are a promising candidate class of dual-acting compounds for treating Parkinson′s disease.
Collapse
Affiliation(s)
- Xuebao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Chao Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yong Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Kaiqi Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Shuangya Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Mangsha Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Luyao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yun Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Faqing Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
40
|
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
|
41
|
Villalba RM, Smith Y. Loss and remodeling of striatal dendritic spines in Parkinson's disease: from homeostasis to maladaptive plasticity? J Neural Transm (Vienna) 2017; 125:431-447. [PMID: 28540422 DOI: 10.1007/s00702-017-1735-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
In Parkinson's disease (PD) patients and animal models of PD, the progressive degeneration of the nigrostriatal dopamine (DA) projection leads to two major changes in the morphology of striatal projection neurons (SPNs), i.e., a profound loss of dendritic spines and the remodeling of axospinous glutamatergic synapses. Striatal spine loss is an early event tightly associated with the extent of striatal DA denervation, but not the severity of parkinsonian motor symptoms, suggesting that striatal spine pruning might be a form of homeostatic plasticity that compensates for the loss of striatal DA innervation and the resulting dysregulation of corticostriatal glutamatergic transmission. On the other hand, the remodeling of axospinous corticostriatal and thalamostriatal glutamatergic synapses might represent a form of late maladaptive plasticity that underlies changes in the strength and plastic properties of these afferents and the resulting increased firing and bursting activity of striatal SPNs in the parkinsonian state. There is also evidence that these abnormal synaptic connections might contribute to the pathophysiology of L-DOPA-induced dyskinesia. Despite the significant advances made in this field over the last thirty years, many controversial issues remain about the striatal SPN subtypes affected, the role of spine changes in the altered activity of SPNs in the parkinsonisn state, and the importance of striatal spine plasticity in the pathophysiology of L-DOPA-induced dyskinesia. In this review, we will examine the current state of knowledge of these issues, discuss the limitations of the animal models used to address some of these questions, and assess the relevance of data from animal models to the human-diseased condition.
Collapse
Affiliation(s)
- Rosa M Villalba
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA. .,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.,Department of Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
42
|
Bentea E, Moore C, Deneyer L, Verbruggen L, Churchill MJ, Hood RL, Meshul CK, Massie A. Plastic changes at corticostriatal synapses predict improved motor function in a partial lesion model of Parkinson’s disease. Brain Res Bull 2017; 130:257-267. [DOI: 10.1016/j.brainresbull.2017.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/16/2017] [Indexed: 12/15/2022]
|
43
|
Henstridge CM, Pickett E, Spires-Jones TL. Synaptic pathology: A shared mechanism in neurological disease. Ageing Res Rev 2016; 28:72-84. [PMID: 27108053 DOI: 10.1016/j.arr.2016.04.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/18/2022]
Abstract
Synaptic proteomes have evolved a rich and complex diversity to allow the exquisite control of neuronal communication and information transfer. It is therefore not surprising that many neurological disorders are associated with alterations in synaptic function. As technology has advanced, our ability to study the anatomical and physiological function of synapses in greater detail has revealed a critical role for both central and peripheral synapses in neurodegenerative disease. Synapse loss has a devastating effect on cellular communication, leading to wide ranging effects such as network disruption within central neural systems and muscle wastage in the periphery. These devastating effects link synaptic pathology to a diverse range of neurological disorders, spanning Alzheimer's disease to multiple sclerosis. This review will highlight some of the current literature on synaptic integrity in animal models of disease and human post-mortem studies. Synaptic changes in normal brain ageing will also be discussed and finally the current and prospective treatments for neurodegenerative disorders will be summarised.
Collapse
Affiliation(s)
| | - Eleanor Pickett
- Centre for Cognitive and Neural Systems, 1 George Square, University of Edinburgh, EH8 9JZ, UK
| | - Tara L Spires-Jones
- Centre for Cognitive and Neural Systems, 1 George Square, University of Edinburgh, EH8 9JZ, UK; Euan MacDonald Centre for Motor Neurone Disease Research, Chancellor's Building, 49 Little France Crescent, University of Edinburgh, EH16 4SB, UK; Centre for Dementia Prevention, University of Edinburgh Kennedy Tower, Royal Edinburgh Hospital, EH10 5HF, UK.
| |
Collapse
|
44
|
Segal M. Dendritic spines: Morphological building blocks of memory. Neurobiol Learn Mem 2016; 138:3-9. [PMID: 27311757 DOI: 10.1016/j.nlm.2016.06.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/22/2016] [Accepted: 06/12/2016] [Indexed: 12/22/2022]
Abstract
The introduction of novel technologies, including high resolution time lapse imaging in behaving animals, molecular modification of the genome and optogenetic control of neuronal excitability have revolutionized the ability to detect subcellular changes in the brain, associated with learning and memory. The sequence of molecular cascades leading to formation, longevity and erasure of memories are being addressed in growing number of studies. Still, major issues concerning the relationship between the morphology and physiology of dendritic spines and memory mechanisms and the functional, neuronal network relevance of such parameters remain unsettled. The present review will summarize recent studies related to the immediate and long lasting changes in density, morphology and function of dendritic spines and their parent neurons following exposure to plasticity-producing stimulation in vivo and in vitro. Standing issues such as the relations between volume/shape and longevity, with respect to different classes of memories in different brain regions will be addressed. These studies indicate that the rules governing the structure/function relations of dendritic spines and memory in the brain are still not conclusive.
Collapse
Affiliation(s)
- Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot 76100, Israel.
| |
Collapse
|
45
|
McIntyre CC, Anderson RW. Deep brain stimulation mechanisms: the control of network activity via neurochemistry modulation. J Neurochem 2016; 139 Suppl 1:338-345. [PMID: 27273305 DOI: 10.1111/jnc.13649] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/04/2016] [Accepted: 04/22/2016] [Indexed: 12/18/2022]
Abstract
Deep brain stimulation (DBS) has revolutionized the clinical care of late-stage Parkinson's disease and shows promise for improving the treatment of intractable neuropsychiatric disorders. However, after over 25 years of clinical experience, numerous questions still remain on the neurophysiological basis for the therapeutic mechanisms of action. At their fundamental core, the general purpose of electrical stimulation therapies in the nervous system are to use the applied electric field to manipulate the opening and closing of voltage-gated sodium channels on neurons, generate stimulation induced action potentials, and subsequently, control the release of neurotransmitters in targeted pathways. Historically, DBS mechanisms research has focused on characterizing the effects of stimulation on neurons and the resulting impact on neuronal network activity. However, when electrodes are placed within the central nervous system, glia are also being directly (and indirectly) influenced by the stimulation. Mounting evidence shows that non-neuronal tissue can play an important role in modulating the neurochemistry changes induced by DBS. The goal of this review is to evaluate how DBS effects on both neuronal and non-neuronal tissue can potentially work together to suppress oscillatory activity (and/or information transfer) between brain regions. These resulting effects of ~ 100 Hz electrical stimulation help explain how DBS can disrupt pathological network activity in the brain and generate therapeutic effects in patients. Deep brain stimulation is an effective clinical technology, but detailed therapeutic mechanisms remain undefined. This review provides an overview of the leading hypotheses, which focus on stimulation-induced disruption of network oscillations and integrates possible roles for non-neuronal tissue in explaining the clinical response to therapeutic stimulation. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Cameron C McIntyre
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA.
| | - Ross W Anderson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Freitas ME, Fox SH. Nondopaminergic treatments for Parkinson's disease: current and future prospects. Neurodegener Dis Manag 2016; 6:249-68. [PMID: 27230697 PMCID: PMC4976881 DOI: 10.2217/nmt-2016-0005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease is primarily caused by dysfunction of dopaminergic neurons, however, nondopaminergic (ND) systems are also involved. ND targets are potentially useful to reduce doses of levodopa or to treat nonlevodopa-responsive symptoms. Recent studies have investigated the role of ND drugs for motor and nonmotor symptoms. Adenosine A2A receptor antagonists, mixed inhibitors of sodium/calcium channels and monoamine oxidase-B have recently been found to improve motor fluctuations. N-methyl-d-aspartate receptor antagonists and serotonin 5HT1B receptor agonists demonstrated benefit in levodopa-induced dyskinesia. Conversely, studies using antiepileptic drugs and adrenoreceptor antagonist had conflicting results. Moreover, metabotropic glutamate receptor antagonists also failed to improve symptoms. The current review summarizes the most recent findings on ND drugs over the last 2 years.
Collapse
Affiliation(s)
- Maria Eliza Freitas
- Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto Western Hospital, 399 Bathurst Street MCL7-412, Toronto, ON M5T 2S8, Canada
| | - Susan H Fox
- Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto Western Hospital, 399 Bathurst Street MCL7-412, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
47
|
Sizemore RJ, Seeger-Armbruster S, Hughes SM, Parr-Brownlie LC. Viral vector-based tools advance knowledge of basal ganglia anatomy and physiology. J Neurophysiol 2016; 115:2124-46. [PMID: 26888111 PMCID: PMC4869490 DOI: 10.1152/jn.01131.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/16/2016] [Indexed: 01/07/2023] Open
Abstract
Viral vectors were originally developed to deliver genes into host cells for therapeutic potential. However, viral vector use in neuroscience research has increased because they enhance interpretation of the anatomy and physiology of brain circuits compared with conventional tract tracing or electrical stimulation techniques. Viral vectors enable neuronal or glial subpopulations to be labeled or stimulated, which can be spatially restricted to a single target nucleus or pathway. Here we review the use of viral vectors to examine the structure and function of motor and limbic basal ganglia (BG) networks in normal and pathological states. We outline the use of viral vectors, particularly lentivirus and adeno-associated virus, in circuit tracing, optogenetic stimulation, and designer drug stimulation experiments. Key studies that have used viral vectors to trace and image pathways and connectivity at gross or ultrastructural levels are reviewed. We explain how optogenetic stimulation and designer drugs used to modulate a distinct pathway and neuronal subpopulation have enhanced our mechanistic understanding of BG function in health and pathophysiology in disease. Finally, we outline how viral vector technology may be applied to neurological and psychiatric conditions to offer new treatments with enhanced outcomes for patients.
Collapse
Affiliation(s)
- Rachel J Sizemore
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Sonja Seeger-Armbruster
- Department of Physiology, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand; and
| | - Stephanie M Hughes
- Department of Biochemistry, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Louise C Parr-Brownlie
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand;
| |
Collapse
|
48
|
Blesa J, Lanciego JL, Obeso JA. Editorial: Parkinson's disease: cell vulnerability and disease progression. Front Neuroanat 2015; 9:125. [PMID: 26441555 PMCID: PMC4569747 DOI: 10.3389/fnana.2015.00125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- Javier Blesa
- HM Hospitales, Centre for Integrative Neuroscience A.C., Hospital Universitario HM Puerta del Sur, Mostoles and CEU San Pablo University Madrid, Spain ; Center for Networked Biomedical Research on Neurodegenerative Diseases, Institute Carlos III Madrid, Spain
| | - Jose L Lanciego
- Center for Networked Biomedical Research on Neurodegenerative Diseases, Institute Carlos III Madrid, Spain ; Center for Applied Medical Research, University of Navarra Medical College Pamplona, Spain
| | - Jose A Obeso
- HM Hospitales, Centre for Integrative Neuroscience A.C., Hospital Universitario HM Puerta del Sur, Mostoles and CEU San Pablo University Madrid, Spain ; Center for Networked Biomedical Research on Neurodegenerative Diseases, Institute Carlos III Madrid, Spain
| |
Collapse
|