51
|
Bore JC, Toth C, Campbell BA, Cho H, Pucci F, Hogue O, Machado AG, Baker KB. Consistent Changes in Cortico-Subthalamic Directed Connectivity Are Associated With the Induction of Parkinsonism in a Chronically Recorded Non-human Primate Model. Front Neurosci 2022; 16:831055. [PMID: 35310095 PMCID: PMC8930827 DOI: 10.3389/fnins.2022.831055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Parkinson’s disease is a neurological disease with cardinal motor signs including bradykinesia and tremor. Although beta-band hypersynchrony in the cortico-basal ganglia network is thought to contribute to disease manifestation, the resulting effects on network connectivity are unclear. We examined local field potentials from a non-human primate across the naïve, mild, and moderate disease states (model was asymmetric, left-hemispheric dominant) and probed power spectral density as well as cortico-cortical and cortico-subthalamic connectivity using both coherence and Granger causality, which measure undirected and directed effective connectivity, respectively. Our network included the left subthalamic nucleus (L-STN), bilateral primary motor cortices (L-M1, R-M1), and bilateral premotor cortices (L-PMC, R-PMC). Results showed two distinct peaks (Peak A at 5–20 Hz, Peak B at 25–45 Hz) across all analyses. Power and coherence analyses showed widespread increases in power and connectivity in both the Peak A and Peak B bands with disease progression. For Granger causality, increases in Peak B connectivity and decreases in Peak A connectivity were associated with the disease. Induction of mild disease was associated with several changes in connectivity: (1) the cortico-subthalamic connectivity in the descending direction (L-PMC to L-STN) decreased in the Peak A range while the reciprocal, ascending connectivity (L-STN to L-PMC) increased in the Peak B range; this may play a role in generating beta-band hypersynchrony in the cortex, (2) both L-M1 to L-PMC and R-M1 to R-PMC causalities increased, which may either be compensatory or a pathologic effect of disease, and (3) a decrease in connectivity occurred from the R-PMC to R-M1. The only significant change seen between mild and moderate disease was increased right cortical connectivity, which may reflect compensation for the left-hemispheric dominant moderate disease state.
Collapse
Affiliation(s)
- Joyce Chelangat Bore
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Carmen Toth
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brett A. Campbell
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Hanbin Cho
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Francesco Pucci
- Center for Neurological Restoration, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
- Department of Neurosurgery, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Olivia Hogue
- Center for Neurological Restoration, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Andre G. Machado
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Center for Neurological Restoration, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
- Department of Neurosurgery, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Kenneth B. Baker
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Center for Neurological Restoration, Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
- *Correspondence: Kenneth B. Baker,
| |
Collapse
|
52
|
Heffernan KS, Rahman K, Smith Y, Galvan A. Characterization of the GfaABC1D Promoter to Selectively Target Astrocytes in the Rhesus Macaque Brain. J Neurosci Methods 2022; 372:109530. [PMID: 35202614 PMCID: PMC8940704 DOI: 10.1016/j.jneumeth.2022.109530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The study of astrocytic functions in non-human primates (NHPs) has been hampered by the lack of genetic tools to selectively target astrocytes. Viral vectors with selective and efficient transduction of astrocytes could be a potent tool to express marker proteins, modulators, or sensors in NHP astrocytes, but the availability of thoroughly characterized astrocytic selective promoter sequences to use in these species remains extremely limited. NEW METHOD We describe the specificity and efficiency of an astrocyte-specific promoter, GfaABC1D in the brain of the rhesus macaque, with emphasis in basal ganglia regions. AAV5-pZac2.1-GfaABC1D-tdTomato was locally injected into the globus pallidus external segment (GPe) and putamen. The extent, efficiency, and specificity of transduction was analyzed with immunohistochemistry at the light and electron microscope levels. RESULTS The GfaABC1D promoter directed the expression of tdTomato in an astrocyte-specific manner in directly or indirectly targeted regions (including both segments of the globus pallidus, putamen, subthalamic nucleus and cortex). COMPARISON WITH EXISTING METHODS Due to its small size, the GfaABC1D promoter is advantageous over other previously used glial fibrillary acidic protein-based promoter sequences, facilitating its use to drive expression of various transgenes in adeno-associated viruses (AAV) or other viral vectors. CONCLUSION GfaABC1D is an efficient promoter that selectively targets astrocytes in the monkey basal ganglia and expands the viral vector toolbox to study astrocytic functions in non-human primates.
Collapse
Affiliation(s)
- Kate S Heffernan
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kazi Rahman
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA; Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Adriana Galvan
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA; Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
53
|
White Matter Microstructural Alterations in Newly Diagnosed Parkinson’s Disease: A Whole-Brain Analysis Using dMRI. Brain Sci 2022; 12:brainsci12020227. [PMID: 35203990 PMCID: PMC8870150 DOI: 10.3390/brainsci12020227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by cardinal motor symptoms and other non-motor symptoms. Studies have investigated various brain areas in PD by detecting white matter alterations using diffusion magnetic resonance imaging processing techniques, which can produce diffusion metrics such as fractional anisotropy and quantitative anisotropy. In this study, we compared the quantitative anisotropy of whole brain regions throughout the subcortical and cortical areas between newly diagnosed PD patients and healthy controls. Additionally, we evaluated the correlations between the quantitative anisotropy of each region and respective neuropsychological test scores to identify the areas most affected by each neuropsychological dysfunction in PD. We found significant quantitative anisotropy differences in several subcortical structures such as the basal ganglia, limbic system, and brain stem as well as in cortical structures such as the temporal lobe, occipital lobe, and insular lobe. Additionally, we found that quantitative anisotropy of some subcortical structures such as the basal ganglia, cerebellum, and brain stem showed the highest correlations with motor dysfunction, whereas cortical structures such as the temporal lobe and occipital lobe showed the highest correlations with olfactory dysfunction in PD. Our study also showed evidence regarding potential neural compensation by revealing higher diffusion metric values in early-stage PD than in healthy controls. We anticipate that our results will improve our understanding of PD’s pathophysiology.
Collapse
|
54
|
Krokidis MG, Exarchos T, Vlamos P. Bioinformatics Approaches for Parkinson's Disease in Clinical Practice: Data-Driven Biomarkers and Pharmacological Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1338:193-198. [PMID: 34973025 DOI: 10.1007/978-3-030-78775-2_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Parkinson's disease is a gradually progressive neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the midbrain area called the substantia nigra pars compacta and cytoplasmic alpha-synuclein-rich inclusions termed Lewy bodies. The etiology and pathogenesis remain incompletely understood. The development of reliable biomarkers for the early and accurate diagnosis, including biochemical, genetic, clinical, and neuroimaging markers, is crucial for unraveling the pathogenic processes of the disease as well as patients' progress surveillance. High-throughput technologies and system biology methodologies can support the identification of potent molecular fingerprints together with the establishment of dynamic network biomarkers. Emphasis is given on multi-omics datasets and dysregulated pathways associated with differentially expressed transcripts, modified protein motifs, and altered metabolic profiles. Although there is no therapy that terminates the neurodegenerative process and dopamine replacement strategy with L-DOPA represents the most effective treatment, numerous therapeutic protocols such as dopamine receptor agonists, MAO-B inhibitors, and cholinesterase inhibitors represent candidate treatments providing at the same time valuable network-based approaches to drug repositioning. Computational methodologies and bioinformatics platforms for visualization, clustering, and validating of molecular and clinical datasets provide important insights into diagnostic processing and therapeutic pipeline.
Collapse
Affiliation(s)
- Marios G Krokidis
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, Corfu, Greece.
| | - Themis Exarchos
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, Corfu, Greece
| | - Panayiotis Vlamos
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, Corfu, Greece
| |
Collapse
|
55
|
Kumar S, Goyal L, Singh S. Tremor and Rigidity in Patients with Parkinson's Disease: Emphasis on Epidemiology, Pathophysiology and Contributing Factors. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:596-609. [PMID: 34620070 DOI: 10.2174/1871527320666211006142100] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/04/2021] [Accepted: 07/03/2021] [Indexed: 06/13/2023]
Abstract
Parkinson's disease (PD) is the second most prominent neurodegenerative movement disorder after Alzheimer's disease, involving 2-3% of the population aged above 65 years. This is mainly triggered by the depletion of dopaminergic neurons located in substantia nigra pars compacta (SNpc) in the region of basal ganglia. At present, diagnosis for symptoms of PD is clinical, contextual, unspecified and therapeutically incomprehensive. Analysis of various causes of PD is essential for an accurate examination of the disease. Among the different causes, such as tremors and rigidity, unresponsiveness to the current treatment approach contributes to mortality. In the present review article, we describe various key factors of pathogenesis and physiology associated with tremors and rigidity necessary for the treatment of PI (postural instability) in patients with PD. Additionally, several reports showing early tremor and rigidity causes, particularly age, cortex lesions, basal ganglia lesions, genetic abnormalities, weakened reflexes, nutrition, fear of fall, and altered biomechanics, have been explored. By summarizing the factors that contribute to the disease, histopathological studies can assess rigidity and tremor in PD. With a clear understanding of the contributing factors, various prospective studies can be done to assess the incidence of rigidity and tremors.
Collapse
Affiliation(s)
- Shivam Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Lav Goyal
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| |
Collapse
|
56
|
Sun S, Zhu Z, He T, Chen F, Wang X, Zhang X, Li M, Li Y, Sun Y, He Q, Li X, Wang M. A study of adeno-associated virus in cortical-thalamostriatal pathway. Brain Res 2021; 1773:147698. [PMID: 34655617 DOI: 10.1016/j.brainres.2021.147698] [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] [Received: 09/01/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022]
Abstract
The cortical-thalamostriatal pathway constitutes the cortico-basal ganglia circuit and plays a critical role in the control of movement. Emerging evidence shows that center median/parafascicular (CM/Pf) neurons are lost in Parkinson's disease (PD) patients with motor deficits and CM/Pf neurons send massive and topographically organized projections to specific regions of the dorsal striatum, but provide only minor inputs to the cerebral cortex. However, anatomical connectivity in the cortical-thalamostriatal pathway are poorly understood at present. In the present study, we used a neural tracing method with adeno-associated virus (AAV) to monitor the cortical-thalamostriatal connectivity in rats. We found that parafascicular nucleus (PF) not only project directly to the striatum but send minor inputs to the cortical regions. It was manifested by green fluorescent protein (GFP) expressing fibers observed in dorsolateral striatum (DLS) and the primary motor cortex (M1) after adeno-associated virus serotype 2/9 (AAV2/9)-GFP injection into PF and GFP expressing cells observed in PF after injection AAV2/retro-GFP into M1. And the PF also receive projections from the DLS and it was demonstrated by GFP expressing fibers in PF after AAV2/9-GFP injection into DLS and GFP expressing cells in DLS after injection AAV2/retro-GFP into PF. Histological and behavioral analysis revealed that AAV vector transduction cause damage in neurons on the injection sites and also damage motor activity of rats suggesting caution in clinical application.
Collapse
Affiliation(s)
- Shuang Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Zhiwei Zhu
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Tianqi He
- Qilu Hospital, Shandong University, Jinan 250014, China
| | - Feiyu Chen
- School of International Education, Qilu University of Technology, Jinan 250014, China
| | - Xiaojun Wang
- The First Hospital Affiliated with Shandong First Medicine University, Jinan 250014, China
| | - Xiao Zhang
- Editorial Department of Journal, Shandong Jianzhu University, Jinan 250014, China
| | - Min Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Yuchuan Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Yue Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Qin He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Xiuhua Li
- The First Hospital Affiliated with Shandong First Medicine University, Jinan 250014, China.
| | - Min Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China.
| |
Collapse
|
57
|
Nakamura KC, Sharott A, Tanaka T, Magill PJ. Input Zone-Selective Dysrhythmia in Motor Thalamus after Dopamine Depletion. J Neurosci 2021; 41:10382-10404. [PMID: 34753740 PMCID: PMC8672689 DOI: 10.1523/jneurosci.1753-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/19/2021] [Accepted: 09/23/2021] [Indexed: 12/01/2022] Open
Abstract
The cerebral cortex, basal ganglia and motor thalamus form circuits important for purposeful movement. In Parkinsonism, basal ganglia neurons often exhibit dysrhythmic activity during, and with respect to, the slow (∼1 Hz) and beta-band (15-30 Hz) oscillations that emerge in cortex in a brain state-dependent manner. There remains, however, a pressing need to elucidate the extent to which motor thalamus activity becomes similarly dysrhythmic after dopamine depletion relevant to Parkinsonism. To address this, we recorded single-neuron and ensemble outputs in the basal ganglia-recipient zone (BZ) and cerebellar-recipient zone (CZ) of motor thalamus in anesthetized male dopamine-intact rats and 6-OHDA-lesioned rats during two brain states, respectively defined by cortical slow-wave activity and activation. Two forms of thalamic input zone-selective dysrhythmia manifested after dopamine depletion: (1) BZ neurons, but not CZ neurons, exhibited abnormal phase-shifted firing with respect to cortical slow oscillations prevalent during slow-wave activity; and (2) BZ neurons, but not CZ neurons, inappropriately synchronized their firing and engaged with the exaggerated cortical beta oscillations arising in activated states. These dysrhythmias were not accompanied by the thalamic hypoactivity predicted by canonical firing rate-based models of circuit organization in Parkinsonism. Complementary recordings of neurons in substantia nigra pars reticulata suggested that their altered activity dynamics could underpin the BZ dysrhythmias. Finally, pharmacological perturbations demonstrated that ongoing activity in the motor thalamus bolsters exaggerated beta oscillations in motor cortex. We conclude that BZ neurons are selectively primed to mediate the detrimental influences of abnormal slow and beta-band rhythms on circuit information processing in Parkinsonism.SIGNIFICANCE STATEMENT Motor thalamus neurons mediate the influences of basal ganglia and cerebellum on the cerebral cortex to govern movement. Chronic depletion of dopamine from the basal ganglia causes some symptoms of Parkinson's disease. Here, we elucidate how dopamine depletion alters the ways motor thalamus neurons engage with two distinct oscillations emerging in cortico-basal ganglia circuits in vivo We discovered that, after dopamine depletion, neurons in the thalamic zone receiving basal ganglia inputs are particularly prone to becoming dysrhythmic, changing the phases and/or synchronization (but not rate) of their action potential firing. This bolsters cortical dysrhythmia. Our results provide important new insights into how aberrant rhythmicity in select parts of motor thalamus could detrimentally affect neural circuit dynamics and behavior in Parkinsonism.
Collapse
Affiliation(s)
- Kouichi C Nakamura
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3TH, United Kingdom
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3TH, United Kingdom
| | - Takuma Tanaka
- Center for Data Science Education and Research, Shiga University, Hikone, Shiga 522-8522, Japan
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3TH, United Kingdom
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, United Kingdom
| |
Collapse
|
58
|
Shim JH, Baek HM. Diffusion Measure Changes of Substantia Nigra Subregions and the Ventral Tegmental Area in Newly Diagnosed Parkinson's Disease. Exp Neurobiol 2021; 30:365-373. [PMID: 34737241 PMCID: PMC8572662 DOI: 10.5607/en21025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Historically, studies have extensively examined the basal ganglia in Parkinson’s disease for specific characteristics that can be observed with medical imaging. One particular methodology used for detecting changes that occur in Parkinson’s disease brains is diffusion tensor imaging, which yields diffusion indices such as fractional anisotropy and radial diffusivity that have been shown to correlate with axonal damage. In this study, we compare the diffusion measures of basal ganglia structures (with substantia nigra divided into subregions, pars compacta, and pars reticula), as well as the diffusion measures of the diffusion tracts that pass through each pair of basal ganglia structures to see if significant differences in diffusion measures can be observed in structures or tracts in newly diagnosed Parkinson’s disease patients. Additionally, we include the ventral tegmental area, a structure connected to various basal ganglia structures affected by dopaminergic neuronal loss and have historically shown significant alterations in Parkinson’s disease, in our analysis. We found significant fractional anisotropy differences in the putamen, and in the diffusion tracts that pass through pairs of both substantia nigra subregions, subthalamic nucleus, parabrachial pigmental nucleus, ventral tegmental area. Additionally, we found significant radial diffusivity differences in diffusion tracts that pass through the parabrachial nucleus, putamen, both substantia nigra subregions, and globus pallidus externa. We were able to find significant diffusion measure differences in structures and diffusion tracts, potentially due to compensatory mechanisms in response to dopaminergic neuronal loss that occurs in newly diagnosed Parkinson’s disease patients.
Collapse
Affiliation(s)
- Jae-Hyuk Shim
- 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
|
59
|
Park JK, Kim SJ. Dual-Task-Based Drum Playing with Rhythmic Cueing on Motor and Attention Control in Patients with Parkinson's Disease: A Preliminary Randomized Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10095. [PMID: 34639396 PMCID: PMC8508067 DOI: 10.3390/ijerph181910095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Although there have been increasing reports regarding the effectiveness of dual-task interventions in rehabilitation, the scope of this research is limited to gross motor movement, such as gait among patients with Parkinson's disease (PD). To expand the dual-task paradigm to upper extremity motor and attention control in PD, drum playing with modulation of musical elements was attempted. The objective of this study was to evaluate the effects of a drum playing intervention with rhythmic cueing on upper extremity motor control and attention control in patients with PD. Twelve participants were randomly assigned to the drum playing intervention with rhythmic cueing group or the control group. The results showed that the drum playing with rhythmic cueing (DPRC) group significantly increased their sustained time of entrainment (45 BPM) and their latency time until entrainment from pretest to posttest. For the DPRC group, the latency time until entrainment was significantly improved, and improvements in cognitive measures were also found. This study shows that DPRC has great potential to improve upper extremity motor control and attention control and supports the development of new interventions that include this technique for rehabilitation in patients with PD.
Collapse
Affiliation(s)
- Jin-Kyoung Park
- Music Therapy Education, Graduate School of Education, Ewha Womans University, Seoul 03760, Korea;
- Laboratory of Brain & Cognitive Sciences for Convergence Medicine, Hallym University College of Medicine, Anyang 14068, Korea
| | - Soo Ji Kim
- Music Therapy Education, Graduate School of Education, Ewha Womans University, Seoul 03760, Korea;
| |
Collapse
|
60
|
Reduced Dopamine Signaling Impacts Pyramidal Neuron Excitability in Mouse Motor Cortex. eNeuro 2021; 8:ENEURO.0548-19.2021. [PMID: 34556558 PMCID: PMC8525657 DOI: 10.1523/eneuro.0548-19.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 08/12/2021] [Accepted: 09/07/2021] [Indexed: 11/21/2022] Open
Abstract
Dopaminergic modulation is essential for the control of voluntary movement; however, the role of dopamine in regulating the neural excitability of the primary motor cortex (M1) is not well understood. Here, we investigated two modes by which dopamine influences the input/output function of M1 neurons. To test the direct regulation of M1 neurons by dopamine, we performed whole-cell recordings of excitatory neurons and measured excitability before and after local, acute dopamine receptor blockade. We then determined whether chronic depletion of dopaminergic input to the entire motor circuit, via a mouse model of Parkinson's disease, was sufficient to shift M1 neuron excitability. We show that D1 receptor (D1R) and D2R antagonism altered subthreshold and suprathreshold properties of M1 pyramidal neurons in a layer-specific fashion. The effects of D1R antagonism were primarily driven by changes to intrinsic properties, while the excitability shifts following D2R antagonism relied on synaptic transmission. In contrast, chronic depletion of dopamine to the motor circuit with 6-hydroxydopamine induced layer-specific synaptic transmission-dependent shifts in M1 neuron excitability that only partially overlapped with the effects of acute D1R antagonism. These results suggest that while acute and chronic changes in dopamine modulate the input/output function of M1 neurons, the mechanisms engaged are distinct depending on the duration and origin of the manipulation. Our study highlights the broad influence of dopamine on M1 excitability by demonstrating the consequences of local and global dopamine depletion on neuronal input/output function.
Collapse
|
61
|
Pimentel JM, Moioli RC, de Araujo MFP, Ranieri CM, Romero RAF, Broz F, Vargas PA. Neuro4PD: An Initial Neurorobotics Model of Parkinson's Disease. Front Neurorobot 2021; 15:640449. [PMID: 34276331 PMCID: PMC8283825 DOI: 10.3389/fnbot.2021.640449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/31/2021] [Indexed: 02/05/2023] Open
Abstract
In this work, we present the first steps toward the creation of a new neurorobotics model of Parkinson's Disease (PD) that embeds, for the first time in a real robot, a well-established computational model of PD. PD mostly affects the modulation of movement in humans. The number of people suffering from this neurodegenerative disease is set to double in the next 15 years and there is still no cure. With the new model we were capable to further explore the dynamics of the disease using a humanoid robot. Results show that the embedded model under both conditions, healthy and parkinsonian, was capable of performing a simple behavioural task with different levels of motor disturbance. We believe that this neurorobotics model is a stepping stone to the development of more sophisticated models that could eventually test and inform new PD therapies and help to reduce and replace animals in research.
Collapse
Affiliation(s)
- Jhielson M. Pimentel
- Edinburgh Centre for Robotics, Heriot-Watt University, Edinburgh, United Kingdom
| | - Renan C. Moioli
- Bioinformatics Multidisciplinary Environment, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | | | - Frank Broz
- Edinburgh Centre for Robotics, Heriot-Watt University, Edinburgh, United Kingdom
| | - Patricia A. Vargas
- Edinburgh Centre for Robotics, Heriot-Watt University, Edinburgh, United Kingdom
| |
Collapse
|
62
|
Huntington TE, Srinivasan R. Adeno-Associated Virus Expression of α-Synuclein as a Tool to Model Parkinson's Disease: Current Understanding and Knowledge Gaps. Aging Dis 2021; 12:1120-1137. [PMID: 34221553 PMCID: PMC8219504 DOI: 10.14336/ad.2021.0517] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder in the aging population and is characterized by a constellation of motor and non-motor symptoms. The abnormal aggregation and spread of alpha-synuclein (α-syn) is thought to underlie the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc), leading to the development of PD. It is in this context that the use of adeno-associated viruses (AAVs) to express a-syn in the rodent midbrain has become a popular tool to model SNc DA neuron loss during PD. In this review, we summarize results from two decades of experiments using AAV-mediated a-syn expression in rodents to model PD. Specifically, we outline aspects of AAV vectors that are particularly relevant to modeling a-syn dysfunction in rodent models of PD such as changes in striatal neurochemistry, a-syn biochemistry, and PD-related behaviors resulting from AAV-mediated a-syn expression in the midbrain. Finally, we discuss the emerging role of astrocytes in propagating a-syn pathology, and point to future directions for employing AAVs as a tool to better understand how astrocytes contribute to a-syn pathology during the development of PD. We envision that lessons learned from two decades of utilizing AAVs to express a-syn in the rodent brain will enable us to develop an optimized set of parameters for gaining a better understanding of how a-syn leads to the development of PD.
Collapse
Affiliation(s)
- Taylor E Huntington
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, 8447 Riverside Pkwy, Bryan, TX 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX 77843, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, 8447 Riverside Pkwy, Bryan, TX 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX 77843, USA
| |
Collapse
|
63
|
Pretzsch CM, Floris DL, Voinescu B, Elsahib M, Mendez MA, Wichers R, Ajram L, Ivin G, Heasman M, Pretzsch E, Williams S, Murphy DGM, Daly E, McAlonan GM. Modulation of striatal functional connectivity differences in adults with and without autism spectrum disorder in a single-dose randomized trial of cannabidivarin. Mol Autism 2021; 12:49. [PMID: 34210360 PMCID: PMC8252312 DOI: 10.1186/s13229-021-00454-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) has a high cost to affected individuals and society, but treatments for core symptoms are lacking. To expand intervention options, it is crucial to gain a better understanding of potential treatment targets, and their engagement, in the brain. For instance, the striatum (caudate, putamen, and nucleus accumbens) plays a central role during development and its (atypical) functional connectivity (FC) may contribute to multiple ASD symptoms. We have previously shown, in the adult autistic and neurotypical brain, the non-intoxicating cannabinoid cannabidivarin (CBDV) alters the balance of striatal 'excitatory-inhibitory' metabolites, which help regulate FC, but the effects of CBDV on (atypical) striatal FC are unknown. METHODS To examine this in a small pilot study, we acquired resting state functional magnetic resonance imaging data from 28 men (15 neurotypicals, 13 ASD) on two occasions in a repeated-measures, double-blind, placebo-controlled study. We then used a seed-based approach to (1) compare striatal FC between groups and (2) examine the effect of pharmacological probing (600 mg CBDV/matched placebo) on atypical striatal FC in ASD. Visits were separated by at least 13 days to allow for drug washout. RESULTS Compared to the neurotypicals, ASD individuals had lower FC between the ventral striatum and frontal and pericentral regions (which have been associated with emotion, motor, and vision processing). Further, they had higher intra-striatal FC and higher putamenal FC with temporal regions involved in speech and language. In ASD, CBDV reduced hyperconnectivity to the neurotypical level. LIMITATIONS Our findings should be considered in light of several methodological aspects, in particular our participant group (restricted to male adults), which limits the generalizability of our findings to the wider and heterogeneous ASD population. CONCLUSION In conclusion, here we show atypical striatal FC with regions commonly associated with ASD symptoms. We further provide preliminary proof of concept that, in the adult autistic brain, acute CBDV administration can modulate atypical striatal circuitry towards neurotypical function. Future studies are required to determine whether modulation of striatal FC is associated with a change in ASD symptoms. TRIAL REGISTRATION clinicaltrials.gov, Identifier: NCT03537950. Registered May 25th, 2018-Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT03537950?term=NCT03537950&draw=2&rank=1 .
Collapse
Affiliation(s)
- Charlotte M. Pretzsch
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
| | - Dorothea L. Floris
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Bogdan Voinescu
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Department of Liaison Psychiatry, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Malka Elsahib
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
| | - Maria A. Mendez
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Robert Wichers
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Department of Psychiatry GGZ Geest, Amsterdam, The Netherlands
| | - Laura Ajram
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Medicines Discovery Catapult, Alderley Park, Alderley Edge, SK10 4TG Cheshire UK
| | - Glynis Ivin
- South London and Maudsley NHS Foundation Trust Pharmacy, London, UK
| | - Martin Heasman
- South London and Maudsley NHS Foundation Trust Pharmacy, London, UK
| | - Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Steven Williams
- Department of Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Declan G. M. Murphy
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
| | - Eileen Daly
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
| | - Gráinne M. McAlonan
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
| |
Collapse
|
64
|
Charroud C, Turella L. Subcortical grey matter changes associated with motor symptoms evaluated by the Unified Parkinson's disease Rating Scale (part III): A longitudinal study in Parkinson's disease. NEUROIMAGE-CLINICAL 2021; 31:102745. [PMID: 34225020 PMCID: PMC8264213 DOI: 10.1016/j.nicl.2021.102745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/26/2021] [Accepted: 06/23/2021] [Indexed: 01/18/2023]
Abstract
Decreased grey matter volume over time suggests a subcortical alteration in PD. Decreased volume in the thalamus may be related to the decline in motor skills. Increased volume in the pallidum may contribute to motor impairment. Structural changes in line with the model of basal ganglia-thalamocortical circuits. VBM and volumetry might capture complementary aspects of structural changes in PD.
Parkinson disease (PD) is characterized by motor deficits related to structural changes in the basal ganglia-thalamocortical circuits. However, it is still unclear the exact nature of the association between grey matter alterations and motor symptoms. Therefore, the aim of our investigation was to identify the subcortical modifications associated with motor symptoms of PD over time - adopting voxel-based morphometry (VBM) and automated volumetry methods. We selected fifty subjects with PD from the Parkinson’s Progression Markers Initiative (PPMI) database, who performed an MRI session at two time points: at baseline (i.e. at maximum 2 years after clinical diagnosis of PD) and after 48 months. Motor symptoms were assessed using the part III of the Unified Parkinson’s Disease Rating Scale at the two time points. Our VBM and volumetric analyses showed a general atrophy in all subcortical regions when comparing baseline with 48 months. These findings confirmed previous observations indicating a subcortical alteration over time in PD. Furthermore, our findings supported the idea that a reduced volume in the thalamus and an increased volume in pallidum may be related to the decline in motor skills. These structural modifications are in accordance with the functional model of the basal ganglia-thalamocortical circuits controlling movements. Moreover, VBM and volumetry provided partially overlapping results, suggesting that these methods might capture complementary aspects of brain degeneration in PD.
Collapse
Affiliation(s)
- Céline Charroud
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Rovereto (TN), Italy.
| | - Luca Turella
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Rovereto (TN), Italy
| |
Collapse
|
65
|
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
|
66
|
Yang YC, Chang FT, Chen JC, Tsai CH, Lin FY, Lu MK. Bereitschaftspotential in Multiple System Atrophy. Front Neurol 2021; 12:608322. [PMID: 34149586 PMCID: PMC8206531 DOI: 10.3389/fneur.2021.608322] [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: 09/25/2020] [Accepted: 05/07/2021] [Indexed: 11/30/2022] Open
Abstract
Objective: Multiple system atrophy (MSA) is a neurodegenerative disorder manifesting as parkinsonism, cerebellar ataxia, and autonomic dysfunction. It is categorized into MSA with predominant parkinsonism (MSA-P) and into MSA with predominant cerebellar ataxia (MSA-C). The pathophysiology of motor control circuitry involvement in MSA subtype is unclear. Bereitschaftspotential (BP) is a feasible clinical tool to measure electroencephalographic activity prior to volitional motions. We recorded BP in patients with MSA-P and MSA-C to investigate their motor cortical preparation and activation for volitional movement. Methods: We included eight patients with MSA-P, eight patients with MSA-C, and eight age-matched healthy controls. BP was recorded during self-paced rapid wrist extension movements. The electroencephalographic epochs were time-locked to the electromyography onset of the voluntary wrist movements. The three groups were compared with respect to the mean amplitudes of early (1,500–500 ms before movement onset) and late (500–0 ms before movement onset) BP. Results: Mean early BP amplitude was non-significantly different between the three groups. Mean late BP amplitude in the two patient groups was significantly reduced in the parietal area contralateral to the movement side compared with that in the healthy control group. In addition, the late BP of the MSA-C group but not the MSA-P group was significantly reduced at the central parietal area compared with that of the healthy control group. Conclusions: Our findings suggest that patients with MSA exhibit motor cortical dysfunction in voluntary movement preparation and activation. The dysfunction can be practicably evaluated using late BP, which represents the cerebello-dentato-thalamo-cortical pathway.
Collapse
Affiliation(s)
- Yi-Chien Yang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Fang-Tzu Chang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Jui-Cheng Chen
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Neurology, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.,Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan.,Ph.D. Program for Translational Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Fu-Yu Lin
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Kuei Lu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Neuroscience and Brain Disease Center, China Medical University Hospital, Taichung, Taiwan.,Ph.D. Program for Translational Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
67
|
Müller-Oehring EM, Hong JY, Hughes RL, Kwon D, Brontë-Stewart HM, Poston KL, Schulte T. Alterations of Brain Signal Oscillations in Older Individuals with HIV Infection and Parkinson's Disease. J Neuroimmune Pharmacol 2021; 16:289-305. [PMID: 32291601 PMCID: PMC7554056 DOI: 10.1007/s11481-020-09914-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/09/2020] [Indexed: 01/01/2023]
Abstract
More than 30 years after the human immunodeficiency virus (HIV) epidemic, HIV patients are now aging due to the advances of antiretroviral therapy. With immunosenescence and the susceptibility of dopamine-rich basal ganglia regions to HIV-related injury, older HIV patients may show neurofunctional deficits similar to patients with Parkinson's disease (PD). We examined the amplitudes of low frequency fluctuations (ALFF) across different frequency bands of the BOLD signal in 30 older HIV-infected individuals, 33 older healthy controls, and 36 PD patients. Participants underwent resting-state fMRI, neuropsychological testing, and a clinical motor exam. HIV patients mainly showed abnormalities in cortical ALFF with reduced prefrontal amplitudes and enhanced sensorimotor and inferior temporal amplitudes. Frontal hypoactivation was overlapping for HIV and PD groups and different from controls. PD patients further exhibited reduced pallidum amplitudes compared to the other groups. In the HIV group, lower pallidum amplitudes were associated with lower CD4+ nadir and CD4+ T cell counts. Abnormalities in ALFF dynamics were largely associated with cognitive and motor functioning in HIV and PD groups. The disruption of neurofunctional frequency dynamics in subcortical-cortical circuits could contribute to the development of cognitive and motor dysfunction and serve as a biomarker for monitoring disease progression with immunosenescence. Graphical Abstract.
Collapse
Affiliation(s)
- Eva M Müller-Oehring
- Neuroscience Program, Biosciences Division, Center for Health Sciences, SRI International, Menlo Park, CA, USA.
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Jui-Yang Hong
- Neuroscience Program, Biosciences Division, Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - Rachel L Hughes
- Department of Psychology, Palo Alto University, Palo Alto, CA, USA
| | - Dongjin Kwon
- Neuroscience Program, Biosciences Division, Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - Helen M Brontë-Stewart
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen L Poston
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tilman Schulte
- Neuroscience Program, Biosciences Division, Center for Health Sciences, SRI International, Menlo Park, CA, USA
- Department of Psychology, Palo Alto University, Palo Alto, CA, USA
| |
Collapse
|
68
|
Brazhnik E, Novikov N, McCoy AJ, Ilieva NM, Ghraib MW, Walters JR. Early decreases in cortical mid-gamma peaks coincide with the onset of motor deficits and precede exaggerated beta build-up in rat models for Parkinson's disease. Neurobiol Dis 2021; 155:105393. [PMID: 34000417 DOI: 10.1016/j.nbd.2021.105393] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 04/27/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022] Open
Abstract
Evidence suggests that exaggerated beta range local field potentials (LFP) in basal ganglia-thalamocortical circuits constitute an important biomarker for feedback for deep brain stimulation in Parkinson's disease patients, although the role of this phenomenon in triggering parkinsonian motor symptoms remains unclear. A useful model for probing the causal role of motor circuit LFP synchronization in motor dysfunction is the unilateral dopamine cell-lesioned rat, which shows dramatic motor deficits walking contralaterally to the lesion but can walk steadily ipsilaterally on a circular treadmill. Within hours after 6-OHDA injection, rats show marked deficits in ipsilateral walking with early loss of significant motor cortex (MCx) LFP peaks in the mid-gamma 41-45 Hz range in the lesioned hemisphere; both effects were reversed by dopamine agonist administration. Increases in MCx and substantia nigra pars reticulata (SNpr) coherence and LFP power in the 29-40 Hz range emerged more gradually over 7 days, although without further progression of walking deficits. Twice-daily chronic dopamine antagonist treatment induced rapid onset of catalepsy and also reduced MCx 41-45 Hz LFP activity at 1 h, with increases in MCx and SNpr 29-40 Hz power/coherence emerging over 7 days, as assessed during periods of walking before the morning treatments. Thus, increases in high beta power in these parkinsonian models emerge gradually and are not linearly correlated with motor deficits. Earlier changes in cortical circuits, reflected in the rapid decreases in MCx LFP mid-gamma LFP activity, may contribute to evolving plasticity supporting increased beta range synchronized activity in basal ganglia-thalamocortical circuits after loss of dopamine receptor stimulation.
Collapse
Affiliation(s)
- Elena Brazhnik
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America
| | - Nikolay Novikov
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America
| | - Alex J McCoy
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America
| | - Neda M Ilieva
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America
| | - Marian W Ghraib
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America
| | - Judith R Walters
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3702, United States of America.
| |
Collapse
|
69
|
Liu A, Bi H, Li Y, Lee S, Cai J, Mi T, Garg S, Kim JL, Zhu M, Chen X, Wang ZJ, McKeown MJ. Galvanic Vestibular Stimulation Improves Subnetwork Interactions in Parkinson's Disease. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6632394. [PMID: 34094040 PMCID: PMC8137296 DOI: 10.1155/2021/6632394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/05/2021] [Indexed: 11/17/2022]
Abstract
Background Activating vestibular afferents via galvanic vestibular stimulation (GVS) has been recently shown to have a number of complex motor effects in Parkinson's disease (PD), but the basis of these improvements is unclear. The evaluation of network-level connectivity changes may provide us with greater insights into the mechanisms of GVS efficacy. Objective To test the effects of different GVS stimuli on brain subnetwork interactions in both health control (HC) and PD groups using fMRI. Methods FMRI data were collected for all participants at baseline (resting state) and under noisy, 1 Hz sinusoidal, and 70-200 Hz multisine GVS. All stimuli were given below sensory threshold, blinding subjects to stimulation. The subnetworks of 15 healthy controls and 27 PD subjects (on medication) were identified in their native space, and their subnetwork interactions were estimated by nonnegative canonical correlation analysis. We then determined if the inferred subnetwork interaction changes were affected by disease and stimulus type and if the stimulus-dependent GVS effects were influenced by demographic features. Results At baseline, interactions with the visual-cerebellar network were significantly decreased in the PD group. Sinusoidal and multisine GVS improved (i.e., made values approaching those seen in HC) subnetwork interactions more effectively than noisy GVS stimuli overall. Worsening disease severity, apathy, depression, impaired cognitive function, and increasing age all limited the beneficial effects of GVS. Conclusions Vestibular stimulation has widespread system-level brain influences and can improve subnetwork interactions in PD in a stimulus-dependent manner, with the magnitude of such effects associating with demographics and disease status.
Collapse
Affiliation(s)
- Aiping Liu
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Huiling Bi
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Yu Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Soojin Lee
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Pacific Parkinson's Research Centre, Vancouver, Canada
| | - Jiayue Cai
- Pacific Parkinson's Research Centre, Vancouver, Canada
| | - Taomian Mi
- Department of Neurology, Neurobiology and Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing Institute of Brain Disorders, Beijing, China
| | - Saurabh Garg
- Pacific Parkinson's Research Centre, Vancouver, Canada
| | - Jowon L. Kim
- Pacific Parkinson's Research Centre, Vancouver, Canada
| | - Maria Zhu
- Pacific Parkinson's Research Centre, Vancouver, Canada
| | - Xun Chen
- Epilepsy Centre, Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Z. Jane Wang
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, Canada
| | - Martin J. McKeown
- Pacific Parkinson's Research Centre, Vancouver, Canada
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, Canada
- Department of Medicine (Neurology), University of British Columbia, Vancouver, Canada
| |
Collapse
|
70
|
Li M, Wang X, Yao X, Wang X, Chen F, Zhang X, Sun S, He F, Jia Q, Guo M, Chen D, Sun Y, Li Y, He Q, Zhu Z, Wang M. Roles of Motor Cortex Neuron Classes in Reach-Related Modulation for Hemiparkinsonian Rats. Front Neurosci 2021; 15:645849. [PMID: 33986639 PMCID: PMC8111217 DOI: 10.3389/fnins.2021.645849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/24/2021] [Indexed: 01/12/2023] Open
Abstract
Disruption of the function of the primary motor cortex (M1) is thought to play a critical role in motor dysfunction in Parkinson's disease (PD). Detailed information regarding the specific aspects of M1 circuits that become abnormal is lacking. We recorded single units and local field potentials (LFPs) of M1 neurons in unilateral 6-hydroxydopamine (6-OHDA) lesion rats and control rats to assess the impact of dopamine (DA) cell loss during rest and a forelimb reaching task. Our results indicated that M1 neurons can be classified into two groups (putative pyramidal neurons and putative interneurons) and that 6-OHDA could modify the activity of different M1 subpopulations to a large extent. Reduced activation of putative pyramidal neurons during inattentive rest and reaching was observed. In addition, 6-OHDA intoxication was associated with an increase in certain LFP frequencies, especially those in the beta range (broadly defined here as any frequency between 12 and 35 Hz), which become pathologically exaggerated throughout cortico-basal ganglia circuits after dopamine depletion. Furthermore, assessment of different spike-LFP coupling parameters revealed that the putative pyramidal neurons were particularly prone to being phase-locked to ongoing cortical oscillations at 12-35 Hz during reaching. Conversely, putative interneurons were neither hypoactive nor synchronized to ongoing cortical oscillations. These data collectively demonstrate a neuron type-selective alteration in the M1 in hemiparkinsonian rats. These alterations hamper the ability of the M1 to contribute to motor conduction and are likely some of the main contributors to motor impairments in PD.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Xuenan Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China.,Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaomeng Yao
- School of Nursing, Qilu Institute of Technology, Jinan, China
| | - Xiaojun Wang
- The First Hospital Affiliated With Shandong First Medicine University, Jinan, China
| | - Feiyu Chen
- School of International Education, Qilu University of Technology, Jinan, China
| | - Xiao Zhang
- Editorial Department of Journal of Shandong Jianzhu University, Jinan, China
| | - Shuang Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Feng He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Qingmei Jia
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Mengnan Guo
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Dadian Chen
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Yue Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Yuchuan Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Qin He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Zhiwei Zhu
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| | - Min Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
| |
Collapse
|
71
|
High-fidelity transmission of high-frequency burst stimuli from peripheral nerve to thalamic nuclei in children with dystonia. Sci Rep 2021; 11:8498. [PMID: 33875779 PMCID: PMC8055985 DOI: 10.1038/s41598-021-88114-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/30/2021] [Indexed: 11/17/2022] Open
Abstract
High-frequency peripheral nerve stimulation has emerged as a noninvasive alternative to thalamic deep brain stimulation for some patients with essential tremor. It is not known whether such techniques might be effective for movement disorders in children, nor is the mechanism and transmission of the peripheral stimuli to central brain structures understood. This study was designed to investigate the fidelity of transmission from peripheral nerves to thalamic nuclei in children with dystonia undergoing deep brain stimulation surgery. The ventralis intermediate (VIM) thalamus nuclei showed a robust evoked response to peripheral high-frequency burst stimulation, with a greatest response magnitude to intra-burst frequencies between 50 and 100 Hz, and reliable but smaller responses up to 170 Hz. The earliest response occurred at 12–15 ms following stimulation onset, suggesting rapid high-fidelity transmission between peripheral nerve and thalamic nuclei. A high-bandwidth, low-latency transmission path from peripheral nerve to VIM thalamus is consistent with the importance of rapid and accurate sensory information for the control of coordination and movement via the cerebello-thalamo-cortical pathway. Our results suggest the possibility of non-invasive modulation of thalamic activity in children with dystonia, and therefore the possibility that a subset of children could have beneficial clinical response without the need for invasive deep brain stimulation.
Collapse
|
72
|
Johnson LA, Aman JE, Yu Y, Escobar Sanabria D, Wang J, Hill M, Dharnipragada R, Patriat R, Fiecas M, Li L, Schrock LE, Cooper SE, Johnson MD, Park MC, Harel N, Vitek JL. High-Frequency Oscillations in the Pallidum: A Pathophysiological Biomarker in Parkinson's Disease? Mov Disord 2021; 36:1332-1341. [PMID: 33847406 DOI: 10.1002/mds.28566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Abnormal oscillatory neural activity in the beta-frequency band (13-35 Hz) is thought to play a role in Parkinson's disease (PD); however, increasing evidence points to alterations in high-frequency ranges (>100 Hz) also having pathophysiological relevance. OBJECTIVES Studies have found that power in subthalamic nucleus (STN) high-frequency oscillations is increased with dopaminergic medication and during voluntary movements, implicating these brain rhythms in normal basal ganglia function. The objective of this study was to investigate whether similar signaling occurs in the internal globus pallidus (GPi), a nucleus increasingly used as a target for deep brain stimulation (DBS) for PD. METHODS Spontaneous and movement-related GPi field potentials were recorded from DBS leads in 5 externalized PD patients on and off dopaminergic medication, as well as from 3 rhesus monkeys before and after the induction of parkinsonism with the neurotoxin 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine. RESULTS In the parkinsonian condition, we identified a prominent oscillatory peak centered at 200-300 Hz that increased during movement. In patients the magnitude of high-frequency oscillation modulation was negatively correlated with bradykinesia. In monkeys, high-frequency oscillations were mostly absent in the naive condition but emerged after the neurotoxin 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine. In patients, spontaneous high-frequency oscillations were significantly attenuated on-medication. CONCLUSIONS Our findings provide evidence in support of the hypothesis that exaggerated, movement-modulated high-frequency oscillations in the GPi are pathophysiological features of PD. These findings suggest that the functional role(s) of high-frequency oscillations may differ between the STN and GPi and motivate additional investigations into their relationship to motor control in normal and diseased states.
Collapse
Affiliation(s)
- Luke A Johnson
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joshua E Aman
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ying Yu
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Jing Wang
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Meghan Hill
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rajiv Dharnipragada
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Remi Patriat
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark Fiecas
- School of Public Health Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura Li
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lauren E Schrock
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Scott E Cooper
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew D Johnson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael C Park
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Noam Harel
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jerrold L Vitek
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
73
|
Tanaka Y, Park IH. Regional specification and complementation with non-neuroectodermal cells in human brain organoids. J Mol Med (Berl) 2021; 99:489-500. [PMID: 33651139 PMCID: PMC8026433 DOI: 10.1007/s00109-021-02051-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/22/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Along with emergence of the organoids, their application in biomedical research has been currently one of the most fascinating themes. For the past few years, scientists have made significant contributions to deriving organoids representing the whole brain and specific brain regions. Coupled with somatic cell reprogramming and CRISPR/Cas9 editing, the organoid technologies were applied for disease modeling and drug screening. The methods to develop organoids further improved for rapid and efficient generation of cerebral organoids. Additionally, refining the methods to develop the regionally specified brain organoids enabled the investigation of development and interaction of the specific brain regions. Recent studies started resolving the issue in the lack of non-neuroectodermal cells in brain organoids, including vascular endothelial cells and microglia, which play fundamental roles in neurodevelopment and are involved in the pathophysiology of acute and chronic neural disorders. In this review, we highlight recent advances of neuronal organoid technologies, focusing on the region-specific brain organoids and complementation with endothelial cells and microglia, and discuss their potential applications to neuronal diseases.
Collapse
Affiliation(s)
- Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA.,Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, QC, H1T 2M4, Canada
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
74
|
Levodopa affects spike and local field synchronisation in the pedunculopontine nucleus of a rat model of Parkinson's disease. Aging (Albany NY) 2021; 13:7314-7329. [PMID: 33639616 PMCID: PMC7993725 DOI: 10.18632/aging.202585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022]
Abstract
The pedunculopontine nucleus (PPN) undergoes significant anatomic and electrophysiological alterations in Parkinson’s disease (PD), severely impacting locomotion. However, the effect of 6-hydroxydopamine (6-OHDA) lesion and levodopa (L-DOPA) therapy on the relationships between spike activities and local field potential (LFP) within the PPN is not well-understood. Synchronisation between the spike activity of individual neurones and LFP of neuronal ensembles is a crucial problem in the pathogenesis of PD. In this study, LFP signals and spikes in the PPN of rats in control, lesioned, and L-DOPA groups were recorded synchronously with a multi-unit electrical signal acquisition system and analysed for their coherence value, spike-field coherence, and phase-lock relationship. The spike-LFP relationship in the PPN was markedly increased in specific frequency bands because of the 6-OHDA lesion but differed depending on the animal locomotion state and neuronal type. L-DOPA had a limited therapeutic effect on the 6-OHDA-induced increase in the coherence value. Our study demonstrates that the PPN spike-LFP relationship is involved in the pathogenesis of PD and is critical for the effects of L-DOPA, providing a basis for the clinical treatment of refractory PD symptoms.
Collapse
|
75
|
Dempsey LM, Kavanagh JJ. Muscarinic receptor blockade causes postcontraction enhancement in corticospinal excitability following maximal contractions. J Neurophysiol 2021; 125:1269-1278. [PMID: 33625939 DOI: 10.1152/jn.00673.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although synaptic transmission in motor pathways can be regulated by neuromodulators, such as acetylcholine, few studies have examined how cholinergic activity affects cortical and spinal motor circuits following muscle contractions of varying intensities. This was a human, double-blinded, placebo-controlled, crossover study. Participants attended two sessions where they were administered either a placebo or 25 mg of promethazine. Electromyography of the abductor digiti minimi (ADM) was measured for all conditions. Motor evoked potentials (MEPs) were obtained via motor cortical transcranial magnetic stimulation (TMS), and F waves were obtained via ulnar nerve electrical stimulation. MEPs and F waves were examined: 1) when the muscle was at rest; 2) after the muscle had been active; and 3) after the muscle had been fatigued. MEPs were unaffected by muscarinic receptor blockade when measurements were recorded from resting muscle or following a 50% isometric maximal voluntary contraction (MVC). However, muscarinic receptor blockade increased MEP area following a 10-s MVC (P = 0.019) and following a fatiguing 60-s MVC (P = 0.040). F wave area and persistence were not affected by promethazine for any muscle contraction condition. Corticospinal excitability was influenced by cholinergic effects when voluntary drive to the muscle was high. Given that spinal motoneurone excitability remained unaffected, it is likely that cholinergic effects are influential within the motor cortex during strong muscle contractions. Future research should evaluate how cholinergic effects alter the relationship between subcortical structures and the motor cortex, as well as brainstem neuromodulatory pathways and spinal motoneurons.NEW & NOTEWORTHY The relationship between motor function and cholinergic circuitry in the central nervous system is complex. Although many studies have approached this issue at the cellular level, few studies have examined cholinergic mechanisms in humans performing muscle contractions. This study demonstrates that blockade of muscarinic acetylcholine receptors enhances motor evoked potentials (elicited with transcranial magnetic stimulation) following strong muscle contractions, but not weak muscle contractions.
Collapse
Affiliation(s)
- Lisa M Dempsey
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Justin J Kavanagh
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
76
|
Aeed F, Cermak N, Schiller J, Schiller Y. Intrinsic Disruption of the M1 Cortical Network in a Mouse Model of Parkinson's Disease. Mov Disord 2021; 36:1565-1577. [PMID: 33606292 DOI: 10.1002/mds.28538] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/14/2020] [Accepted: 01/15/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) disrupts motor performance by affecting the basal ganglia system. Yet, despite the critical position of the primary motor cortex in linking basal ganglia computations with motor performance, its contribution to motor disability in PD is largely unknown. The objective of this study was to characterize the role of the primary motor cortex in PD-related motor disability. METHODS Two-photon calcium imaging and optogenetic stimulation of primary motor cortex neurons was done during performance of a dexterous reach-to-grasp motor task in control and 6-hydroxydopamine-induced PD mice. RESULTS Experimental PD disrupted performance of the reach-to-grasp motor task and especially initiation of the task, which was partially restored by optogenetic activation of the primary motor cortex. Two-photon calcium imaging during task performance revealed experimental-PD affected the primary motor cortex in a cell-type-specific manner. It suppressed activation of output layer 5 pyramidal tract neurons, with greater effects on freeze versus nonfreeze trials. In contrast, it did not attenuate the initial movement-related activation response of layer 2/3 pyramidal neurons while diminishing the late inhibitory phase of their response. At the network level, experimental PD disrupted movement-related population dynamics of the layer 5 pyramidal tract network while almost not affecting the dynamics of the layer 2/3 neuronal population. It also disrupted short- and long-term robustness and stability of the pyramidal tract subnetwork, with reduced intertrial temporal accuracy and diminished reproducibility of motor parameter encoding and temporal recruitment of the output pyramidal tract neurons over repeated daily sessions. CONCLUSIONS Experimental PD disrupts both external driving and intrinsic properties of the primary motor cortex. Motor disability in experimental PD results primarily from the inability to generate robust and stable output motor sequences in the parkinsonian primary motor cortex output layer 5 pyramidal tract subnetwork. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Fadi Aeed
- The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nathan Cermak
- The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jackie Schiller
- The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yitzhak Schiller
- The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.,Department of Neurology, Rambam Medical Center, Haifa, Israel
| |
Collapse
|
77
|
Sedaghat K, Gundlach AL, Finkelstein DI. Analysis of morphological and neurochemical changes in subthalamic nucleus neurons in response to a unilateral 6-OHDA lesion of the substantia nigra in adult rats. IBRO Neurosci Rep 2021; 10:96-103. [PMID: 33842916 PMCID: PMC8019994 DOI: 10.1016/j.ibneur.2021.01.002] [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: 10/12/2020] [Revised: 12/28/2020] [Accepted: 01/12/2021] [Indexed: 11/29/2022] Open
Abstract
Background Subthalamic nucleus (STN) neurons undergo changes in their pattern of activity and morphology during the clinical course of Parkinson’s disease (PD). Striatal dopamine depletion and hyperactivity of neurons in the parafascicular nucleus (Pf) of the intralaminar thalamus are predicted to contribute to the STN changes. Objective This study investigated possible morphological and neurochemical changes in STN neurons in a rat model of unilateral, nigral dopamine neuron loss, in relation to previously documented alterations in Pf neurons. Methods Male Sprague-Dawley rats received a unilateral injection of 6-hydroxydopamine (6-OHDA) into the substantia nigra pars compacta (SNpc). Rats were randomly divided into two groups (6/group) for study at 1 and 5 months by post-treatment. The extent of SNpc dopamine neuron damage was assessed in an amphetamine-induced rotation test and postmortem assessment of tyrosine hydroxylase mRNA levels using in situ hybridization histochemistry. Neural cross-sectional measurements and assessment of vesicular glutamate transporter-2 (vGlut2) mRNA levels were performed to measure the impact on neurons in the STN. Results A unilateral SNpc dopaminergic neuron lesion significantly decreased the cross-sectional area of STN neurons ipsilateral to the lesion, at 1 month (P < 0.05) and 5 months (P < 0.01) post-lesion, while bilateral vGlut2 mRNA levels in STN neurons were unaltered. Conclusions Decreased size of STN neurons in the presence of sustained vGlut2 mRNA levels following a unilateral SNpc 6-OHDA lesion, indicate altered STN physiology. This study presents further details of changes within the STN, coincident with observed alterations in Pf neurons and behaviour. Data availability The data associated with the findings of this study are available from the corresponding author upon request.
Collapse
Affiliation(s)
- Katayoun Sedaghat
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| |
Collapse
|
78
|
Patricio F, Morales-Andrade AA, Patricio-Martínez A, Limón ID. Cannabidiol as a Therapeutic Target: Evidence of its Neuroprotective and Neuromodulatory Function in Parkinson's Disease. Front Pharmacol 2020; 11:595635. [PMID: 33384602 PMCID: PMC7770114 DOI: 10.3389/fphar.2020.595635] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
The phytocannabinoids of Cannabis sativa L. have, since ancient times, been proposed as a pharmacological alternative for treating various central nervous system (CNS) disorders. Interestingly, cannabinoid receptors (CBRs) are highly expressed in the basal ganglia (BG) circuit of both animals and humans. The BG are subcortical structures that regulate the initiation, execution, and orientation of movement. CBRs regulate dopaminergic transmission in the nigro-striatal pathway and, thus, the BG circuit also. The functioning of the BG is affected in pathologies related to movement disorders, especially those occurring in Parkinson’s disease (PD), which produces motor and non-motor symptoms that involving GABAergic, glutamatergic, and dopaminergic neural networks. To date, the most effective medication for PD is levodopa (l-DOPA); however, long-term levodopa treatment causes a type of long-term dyskinesias, l-DOPA-induced dyskinesias (LIDs). With neuromodulation offering a novel treatment strategy for PD patients, research has focused on the endocannabinoid system (ECS), as it participates in the physiological neuromodulation of the BG in order to control movement. CBRs have been shown to inhibit neurotransmitter release, while endocannabinoids (eCBs) play a key role in the synaptic regulation of the BG. In the past decade, cannabidiol (CBD), a non-psychotropic phytocannabinoid, has been shown to have compensatory effects both on the ECS and as a neuromodulator and neuroprotector in models such as 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and reserpine, as well as other PD models. Although the CBD-induced neuroprotection observed in animal models of PD has been attributed to the activation of the CB1 receptor, recent research conducted at a molecular level has proposed that CBD is capable of activating other receptors, such as CB2 and the TRPV-1 receptor, both of which are expressed in the dopaminergic neurons of the nigro-striatal pathway. These findings open new lines of scientific inquiry into the effects of CBD at the level of neural communication. Cannabidiol activates the PPARγ, GPR55, GPR3, GPR6, GPR12, and GPR18 receptors, causing a variety of biochemical, molecular, and behavioral effects due to the broad range of receptors it activates in the CNS. Given the low number of pharmacological treatment alternatives for PD currently available, the search for molecules with the therapeutic potential to improve neuronal communication is crucial. Therefore, the investigation of CBD and the mechanisms involved in its function is required in order to ascertain whether receptor activation could be a treatment alternative for both PD and LID.
Collapse
Affiliation(s)
- Felipe Patricio
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alan Axel Morales-Andrade
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.,Facultad De Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
79
|
A dynamical model for the basal ganglia-thalamo-cortical oscillatory activity and its implications in Parkinson's disease. Cogn Neurodyn 2020; 15:693-720. [PMID: 34367369 PMCID: PMC8286922 DOI: 10.1007/s11571-020-09653-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/27/2022] Open
Abstract
We propose to investigate brain electrophysiological alterations associated with Parkinson’s disease through a novel adaptive dynamical model of the network of the basal ganglia, the cortex and the thalamus. The model uniquely unifies the influence of dopamine in the regulation of the activity of all basal ganglia nuclei, the self-organised neuronal interdependent activity of basal ganglia-thalamo-cortical circuits and the generation of subcortical background oscillations. Variations in the amount of dopamine produced in the neurons of the substantia nigra pars compacta are key both in the onset of Parkinson’s disease and in the basal ganglia action selection. We model these dopamine-induced relationships, and Parkinsonian states are interpreted as spontaneous emergent behaviours associated with different rhythms of oscillatory activity patterns of the basal ganglia-thalamo-cortical network. These results are significant because: (1) the neural populations are built upon single-neuron models that have been robustly designed to have eletrophysiologically-realistic responses, and (2) our model distinctively links changes in the oscillatory activity in subcortical structures, dopamine levels in the basal ganglia and pathological synchronisation neuronal patterns compatible with Parkinsonian states, this still remains an open problem and is crucial to better understand the progression of the disease.
Collapse
|
80
|
Autophagy and Redox Homeostasis in Parkinson's: A Crucial Balancing Act. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8865611. [PMID: 33224433 PMCID: PMC7671810 DOI: 10.1155/2020/8865611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated primarily from endogenous biochemical reactions in mitochondria, endoplasmic reticulum (ER), and peroxisomes. Typically, ROS/RNS correlate with oxidative damage and cell death; however, free radicals are also crucial for normal cellular functions, including supporting neuronal homeostasis. ROS/RNS levels influence and are influenced by antioxidant systems, including the catabolic autophagy pathways. Autophagy is an intracellular lysosomal degradation process by which invasive, damaged, or redundant cytoplasmic components, including microorganisms and defunct organelles, are removed to maintain cellular homeostasis. This process is particularly important in neurons that are required to cope with prolonged and sustained operational stress. Consequently, autophagy is a primary line of protection against neurodegenerative diseases. Parkinson's is caused by the loss of midbrain dopaminergic neurons (mDANs), resulting in progressive disruption of the nigrostriatal pathway, leading to motor, behavioural, and cognitive impairments. Mitochondrial dysfunction, with associated increases in oxidative stress, and declining proteostasis control, are key contributors during mDAN demise in Parkinson's. In this review, we analyse the crosstalk between autophagy and redoxtasis, including the molecular mechanisms involved and the detrimental effect of an imbalance in the pathogenesis of Parkinson's.
Collapse
|
81
|
Primary motor cortex in Parkinson's disease: Functional changes and opportunities for neurostimulation. Neurobiol Dis 2020; 147:105159. [PMID: 33152506 DOI: 10.1016/j.nbd.2020.105159] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023] Open
Abstract
Movement abnormalities of Parkinson's disease (PD) arise from disordered neural activity in multiple interconnected brain structures. The planning and execution of movement requires recruitment of a heterogeneous collection of pyramidal projection neurons in the primary motor cortex (M1). The neural representations of movement in M1 single-cell and field potential recordings are directly and indirectly influenced by the midbrain dopaminergic neurons that degenerate in PD. This review examines M1 functional alterations in PD as uncovered by electrophysiological recordings and neurostimulation studies in patients and experimental animal models. Dysfunction of the parkinsonian M1 depends on the severity and/or duration of dopamine-depletion and the species examined, and is expressed as alterations in movement-related firing dynamics; functional reorganisation of local circuits; and changes in field potential beta oscillations. Neurostimulation methods that modulate M1 activity directly (e.g., transcranial magnetic stimulation) or indirectly (subthalamic nucleus deep brain stimulation) improve motor function in PD patients, showing that targeted neuromodulation of M1 is a realistic therapy. We argue that the therapeutic profile of M1 neurostimulation is likely to be greatly enhanced with alternative technologies that permit cell-type specific control and incorporate feedback from electrophysiological biomarkers measured locally.
Collapse
|
82
|
Parkinson disease and the gut: new insights into pathogenesis and clinical relevance. Nat Rev Gastroenterol Hepatol 2020; 17:673-685. [PMID: 32737460 DOI: 10.1038/s41575-020-0339-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
The classic view portrays Parkinson disease (PD) as a motor disorder resulting from loss of substantia nigra pars compacta dopaminergic neurons. Multiple studies, however, describe prodromal, non-motor dysfunctions that affect the quality of life of patients who subsequently develop PD. These prodromal dysfunctions comprise a wide array of gastrointestinal motility disorders including dysphagia, delayed gastric emptying and chronic constipation. The histological hallmark of PD - misfolded α-synuclein aggregates that form Lewy bodies and neurites - is detected in the enteric nervous system prior to clinical diagnosis, suggesting that the gastrointestinal tract and its neural (vagal) connection to the central nervous system could have a major role in disease aetiology. This Review provides novel insights on the pathogenesis of PD, including gut-to-brain trafficking of α-synuclein as well as the newly discovered nigro-vagal pathway, and highlights how vagal connections from the gut could be the conduit by which ingested environmental pathogens enter the central nervous system and ultimately induce, or accelerate, PD progression. The pathogenic potential of various environmental neurotoxicants and the suitability and translational potential of experimental animal models of PD will be highlighted and appraised. Finally, the clinical manifestations of gastrointestinal involvement in PD and medications will be discussed briefly.
Collapse
|
83
|
Hsu SY, Yeh LR, Chen TB, Du WC, Huang YH, Twan WH, Lin MC, Hsu YH, Wu YC, Chen HY. Classification of the Multiple Stages of Parkinson's Disease by a Deep Convolution Neural Network Based on 99mTc-TRODAT-1 SPECT Images. Molecules 2020; 25:E4792. [PMID: 33086589 PMCID: PMC7587595 DOI: 10.3390/molecules25204792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/03/2022] Open
Abstract
Single photon emission computed tomography (SPECT) has been employed to detect Parkinson's disease (PD). However, analysis of the SPECT PD images was mostly based on the region of interest (ROI) approach. Due to limited size of the ROI, especially in the multi-stage classification of PD, this study utilizes deep learning methods to establish a multiple stages classification model of PD. In the retrospective study, the 99mTc-TRODAT-1 was used for brain SPECT imaging. A total of 202 cases were collected, and five slices were selected for analysis from each subject. The total number of images was thus 1010. According to the Hoehn and Yahr Scale standards, all the cases were divided into healthy, early, middle, late four stages, and HYS I~V six stages. Deep learning is compared with five convolutional neural networks (CNNs). The input images included grayscale and pseudo color of two types. The training and validation sets were 70% and 30%. The accuracy, recall, precision, F-score, and Kappa values were used to evaluate the models' performance. The best accuracy of the models based on grayscale and color images in four and six stages were 0.83 (AlexNet), 0.85 (VGG), 0.78 (DenseNet) and 0.78 (DenseNet).
Collapse
Affiliation(s)
- Shih-Yen Hsu
- Department of Medical Imaging and Radiological Science, I-Shou University, No. 8, Yida Road., Jiao-su Village Yan-chao District, Kaohsiung City 82445, Taiwan; (S.-Y.H.); (L.-R.Y.); (T.-B.C.); (Y.-H.H.)
| | - Li-Ren Yeh
- Department of Medical Imaging and Radiological Science, I-Shou University, No. 8, Yida Road., Jiao-su Village Yan-chao District, Kaohsiung City 82445, Taiwan; (S.-Y.H.); (L.-R.Y.); (T.-B.C.); (Y.-H.H.)
- Department of Anesthesiology, E-DA Cancer Hospital, I-Shou University, No.1, Yida Road, Jiao-su Village, Yan-chao District, Kaohsiung City 82445, Taiwan
| | - Tai-Been Chen
- Department of Medical Imaging and Radiological Science, I-Shou University, No. 8, Yida Road., Jiao-su Village Yan-chao District, Kaohsiung City 82445, Taiwan; (S.-Y.H.); (L.-R.Y.); (T.-B.C.); (Y.-H.H.)
| | - Wei-Chang Du
- Department of Information Engineering, I-Shou University, No.1, Sec. 1, Syuecheng Road., Dashu District, Kaohsiung 84001, Taiwan;
| | - Yung-Hui Huang
- Department of Medical Imaging and Radiological Science, I-Shou University, No. 8, Yida Road., Jiao-su Village Yan-chao District, Kaohsiung City 82445, Taiwan; (S.-Y.H.); (L.-R.Y.); (T.-B.C.); (Y.-H.H.)
| | - Wen-Hung Twan
- Department of Life Sciences, National Taitung University, No.369, Sec. 2, University Road, Taitung 95092, Taiwan;
| | - Ming-Chia Lin
- Department of Nuclear Medicine, E-DA Hospital, I-Shou University, No.1, Yida Rd, Jiao-su Village, Yan-chao District, Kaohsiung 82445, Taiwan; (M.-C.L.); (Y.-H.H.)
| | - Yun-Hsuan Hsu
- Department of Nuclear Medicine, E-DA Hospital, I-Shou University, No.1, Yida Rd, Jiao-su Village, Yan-chao District, Kaohsiung 82445, Taiwan; (M.-C.L.); (Y.-H.H.)
| | - Yi-Chen Wu
- Department of Medical Imaging and Radiological Science, I-Shou University, No. 8, Yida Road., Jiao-su Village Yan-chao District, Kaohsiung City 82445, Taiwan; (S.-Y.H.); (L.-R.Y.); (T.-B.C.); (Y.-H.H.)
- Department of Information Engineering, I-Shou University, No.1, Sec. 1, Syuecheng Road., Dashu District, Kaohsiung 84001, Taiwan;
- Department of Nuclear Medicine, E-DA Hospital, I-Shou University, No.1, Yida Rd, Jiao-su Village, Yan-chao District, Kaohsiung 82445, Taiwan; (M.-C.L.); (Y.-H.H.)
| | - Huei-Yung Chen
- Department of Nuclear Medicine, E-DA Hospital, I-Shou University, No.1, Yida Rd, Jiao-su Village, Yan-chao District, Kaohsiung 82445, Taiwan; (M.-C.L.); (Y.-H.H.)
| |
Collapse
|
84
|
Hernandez-Martin E, Arguelles E, Deshpande R, Sanger TD. Evoked Potentials During Peripheral Stimulation Confirm Electrode Location in Thalamic Subnuclei in Children With Secondary Dystonia. J Child Neurol 2020; 35:799-807. [PMID: 32567481 DOI: 10.1177/0883073820931970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Deep brain stimulation is an elective surgical intervention that improves the function and quality of life in children with dystonia and other movement disorders. Both basal ganglia and thalamic nuclei have been found to be relevant targets for treatment of dystonia in children, including the ventral intermediate nucleus of the thalamus, in which stimulation can control dystonic spasms. Electrophysiological confirmation of correct electrode location within the ventralis intermediate nucleus is thus important for the success of the surgical outcome. The present work shows the evoked potentials response during contralateral median-nerve stimulation at the wrist at low frequency (9 Hz) provides physiological evidence of the electrode's localization within the thalamus. We show the correlation between evoked potentials and magnetic resonance imaging (MRI) and computed tomography (CT) in 14 children undergoing implantation of deep brain stimulation electrodes for secondary dystonia. High fidelity and reproducibility of our results provides a new approach to ensure the electrode localization in the thalamic subnuclei.
Collapse
Affiliation(s)
- Estefania Hernandez-Martin
- Department of Electrical Engineering and Computer Science, 8788University of California, Irvine, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, 5116University of Southern California, Los Angeles, CA, USA
| | - Enrique Arguelles
- Department of Electrical Engineering and Computer Science, 8788University of California, Irvine, CA, USA
| | - Ruta Deshpande
- Department of Electrical Engineering and Computer Science, 8788University of California, Irvine, CA, USA
| | - Terence D Sanger
- Department of Electrical Engineering and Computer Science, 8788University of California, Irvine, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, 5116University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
85
|
Mueller K, Urgošík D, Ballarini T, Holiga Š, Möller HE, Růžička F, Roth J, Vymazal J, Schroeter ML, Růžička E, Jech R. Differential effects of deep brain stimulation and levodopa on brain activity in Parkinson's disease. Brain Commun 2020; 2:fcaa005. [PMID: 32954278 PMCID: PMC7425344 DOI: 10.1093/braincomms/fcaa005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/21/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Levodopa is the first-line treatment for Parkinson’s disease, although the precise mechanisms mediating its efficacy remain elusive. We aimed to elucidate treatment effects of levodopa on brain activity during the execution of fine movements and to compare them with deep brain stimulation of the subthalamic nuclei. We studied 32 patients with Parkinson’s disease using functional MRI during the execution of finger-tapping task, alternating epochs of movement and rest. The task was performed after withdrawal and administration of a single levodopa dose. A subgroup of patients (n = 18) repeated the experiment after electrode implantation with stimulator on and off. Investigating levodopa treatment, we found a significant interaction between both factors of treatment state (off, on) and experimental task (finger tapping, rest) in bilateral putamen, but not in other motor regions. Specifically, during the off state of levodopa medication, activity in the putamen at rest was higher than during tapping. This represents an aberrant activity pattern probably indicating the derangement of basal ganglia network activity due to the lack of dopaminergic input. Levodopa medication reverted this pattern, so that putaminal activity during finger tapping was higher than during rest, as previously described in healthy controls. Within-group comparison with deep brain stimulation underlines the specificity of our findings with levodopa treatment. Indeed, a significant interaction was observed between treatment approach (levodopa, deep brain stimulation) and treatment state (off, on) in bilateral putamen. Our functional MRI study compared for the first time the differential effects of levodopa treatment and deep brain stimulation on brain motor activity. We showed modulatory effects of levodopa on brain activity of the putamen during finger movement execution, which were not observed with deep brain stimulation.
Collapse
Affiliation(s)
- Karsten Mueller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Dušan Urgošík
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Stereotactic and Radiation Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Tommaso Ballarini
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Štefan Holiga
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Filip Růžička
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Stereotactic and Radiation Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Jan Roth
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Stereotactic and Radiation Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Josef Vymazal
- Department of Stereotactic and Radiation Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Matthias L Schroeter
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Evžen Růžička
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Robert Jech
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Stereotactic and Radiation Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| |
Collapse
|
86
|
Troncoso-Escudero P, Sepulveda D, Pérez-Arancibia R, Parra AV, Arcos J, Grunenwald F, Vidal RL. On the Right Track to Treat Movement Disorders: Promising Therapeutic Approaches for Parkinson's and Huntington's Disease. Front Aging Neurosci 2020; 12:571185. [PMID: 33101007 PMCID: PMC7497570 DOI: 10.3389/fnagi.2020.571185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Movement disorders are neurological conditions in which patients manifest a diverse range of movement impairments. Distinct structures within the basal ganglia of the brain, an area involved in movement regulation, are differentially affected for every disease. Among the most studied movement disorder conditions are Parkinson's (PD) and Huntington's disease (HD), in which the deregulation of the movement circuitry due to the loss of specific neuronal populations in basal ganglia is the underlying cause of motor symptoms. These symptoms are due to the loss principally of dopaminergic neurons of the substantia nigra (SN) par compacta and the GABAergic neurons of the striatum in PD and HD, respectively. Although these diseases were described in the 19th century, no effective treatment can slow down, reverse, or stop disease progression. Available pharmacological therapies have been focused on preventing or alleviating motor symptoms to improve the quality of life of patients, but these drugs are not able to mitigate the progressive neurodegeneration. Currently, considerable therapeutic advances have been achieved seeking a more efficacious and durable therapeutic effect. Here, we will focus on the new advances of several therapeutic approaches for PD and HD, starting with the available pharmacological treatments to alleviate the motor symptoms in both diseases. Then, we describe therapeutic strategies that aim to restore specific neuronal populations or their activity. Among the discussed strategies, the use of Neurotrophic factors (NTFs) and genetic approaches to prevent the neuronal loss in these diseases will be described. We will highlight strategies that have been evaluated in both Parkinson's and Huntington's patients, and also the ones with strong preclinical evidence. These current therapeutic techniques represent the most promising tools for the safe treatment of both diseases, specifically those aimed to avoid neuronal loss during disease progression.
Collapse
Affiliation(s)
- Paulina Troncoso-Escudero
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Denisse Sepulveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rodrigo Pérez-Arancibia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Alejandra V. Parra
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Javiera Arcos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Felipe Grunenwald
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rene L. Vidal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| |
Collapse
|
87
|
Maith O, Villagrasa Escudero F, Dinkelbach HÜ, Baladron J, Horn A, Irmen F, Kühn AA, Hamker FH. A computational model‐based analysis of basal ganglia pathway changes in Parkinson’s disease inferred from resting‐state fMRI. Eur J Neurosci 2020; 53:2278-2295. [DOI: 10.1111/ejn.14868] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Oliver Maith
- Department of Computer Science Chemnitz University of Technology Chemnitz Germany
| | | | - Helge Ülo Dinkelbach
- Department of Computer Science Chemnitz University of Technology Chemnitz Germany
| | - Javier Baladron
- Department of Computer Science Chemnitz University of Technology Chemnitz Germany
| | - Andreas Horn
- Movement Disorders and Neuromodulation Unit, Department for Neurology Charité–University Medicine Berlin Berlin Germany
| | - Friederike Irmen
- Movement Disorders and Neuromodulation Unit, Department for Neurology Charité–University Medicine Berlin Berlin Germany
| | - Andrea A. Kühn
- Movement Disorders and Neuromodulation Unit, Department for Neurology Charité–University Medicine Berlin Berlin Germany
| | - Fred H. Hamker
- Department of Computer Science Chemnitz University of Technology Chemnitz Germany
| |
Collapse
|
88
|
Pol S, Temel Y, Jahanshahi A. A Custom Made Electrode Construct and Reliable Implantation Method That Allows for Long-Term Bilateral Deep Brain Stimulation in Mice. Neuromodulation 2020; 24:212-219. [PMID: 32385967 PMCID: PMC7984026 DOI: 10.1111/ner.13165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 03/20/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Objectives The underlying mechanisms behind the therapeutic and side effects of deep brain stimulation (DBS) need further investigation. The utilization of transgenic mouse lines is a suitable approach to better understand the cellular and network effects of DBS. However, not many bilateral DBS studies have been conducted in mice. This might be due to a lack of commercially available bilateral DBS constructs. Materials and Methods We developed an approach to perform repetitive long‐term DBS in freely moving mice. In this study, we implanted an in‐house custom‐made DBS construct containing two bipolar concentric electrodes to target the subthalamic nucleus (STN) bilaterally. Subsequently, we stimulated half of the animals with clinically relevant parameters three to five times a week with a duration of 20 min for ten weeks. Several behavioral tests were conducted of which the open field test (OFT) is shown to validate the reliability of this electrode construct and implantation method. Furthermore, we performed fiber photometry measurements to show the acute effect of STN‐DBS on serotonin network activity in the dorsal raphe nucleus. Results Repetitive DBS and long‐term behavioral testing were performed without complications. STN‐DBS resulted in an increase of the distance traveled in the OFT and a reduction of calcium levels in serotonergic neurons of the dorsal raphe nucleus. None of the mice had lost their electrodes and postmortem evaluation of the tissue showed accurate targeting of the STN without excessive gliosis. Conclusion The DBS electrode construct and implantation method described can be used for long‐term DBS studies to further investigate the mechanisms underlying DBS.
Collapse
Affiliation(s)
- Sylvana Pol
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ali Jahanshahi
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
89
|
Devergnas A, Caiola M, Pittard D, Wichmann T. Cortical Phase-Amplitude Coupling in a Progressive Model of Parkinsonism in Nonhuman Primates. Cereb Cortex 2020; 29:167-177. [PMID: 29190329 DOI: 10.1093/cercor/bhx314] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease is associated with abnormal oscillatory electrical activities of neurons and neuronal ensembles throughout the basal ganglia-thalamocortical network. It has recently been documented in patients with advanced parkinsonism that the amplitude of gamma-band oscillations (50-200 Hz) in electrocorticogram recordings from the primary motor cortex is abnormally coupled to the phase of beta band oscillations within the same signals. It is not known when in the course of the disease the abnormal phase-amplitude coupling (PAC) arises, and whether it is influenced by arousal or prior exposure to dopaminergic medications. To address these issues, we analyzed the relationship between the severity of parkinsonian motor signs and the extent of PAC in a progressive model of parkinsonism, using primates that were not exposed to levodopa prior to testing. PAC was measured in electrocorticogram signals from the primary motor cortex and the supplementary motor area in 3 monkeys that underwent weekly injections of small doses of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rendering them progressively parkinsonian. We found that parkinsonism was associated with increased coupling between the phase of low-frequency (4-10 Hz) oscillations and the amplitude of oscillations in the high gamma band (50-150 Hz). These changes only reached significance when the animals became fully parkinsonian. The increased PAC was normalized after levodopa treatment. We also found a similar increase in PAC during sleep, even in normal animals. The identified PAC was independent of concomitant changes in spectral power in the 2.9-9.8Hz or 49.8-150.4 Hz ranges. We conclude that PAC is predominately a sign of advanced parkinsonism, and is, thus, not essential for the development of parkinsonism. However, increased PAC appears to correlate with the severity of fully developed parkinsonism.
Collapse
Affiliation(s)
- Annaelle Devergnas
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA.,Morris K. Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| | - M Caiola
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Morris K. Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| | - D Pittard
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Morris K. Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| | - T Wichmann
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA.,Morris K. Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| |
Collapse
|
90
|
Mulcahy G, Atwood B, Kuznetsov A. Basal ganglia role in learning rewarded actions and executing previously learned choices: Healthy and diseased states. PLoS One 2020; 15:e0228081. [PMID: 32040519 PMCID: PMC7010262 DOI: 10.1371/journal.pone.0228081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 01/07/2020] [Indexed: 01/06/2023] Open
Abstract
The basal ganglia (BG) is a collection of nuclei located deep beneath the cerebral cortex that is involved in learning and selection of rewarded actions. Here, we analyzed BG mechanisms that enable these functions. We implemented a rate model of a BG-thalamo-cortical loop and simulated its performance in a standard action selection task. We have shown that potentiation of corticostriatal synapses enables learning of a rewarded option. However, these synapses became redundant later as direct connections between prefrontal and premotor cortices (PFC-PMC) were potentiated by Hebbian learning. After we switched the reward to the previously unrewarded option (reversal), the BG was again responsible for switching to the new option. Due to the potentiated direct cortical connections, the system was biased to the previously rewarded choice, and establishing the new choice required a greater number of trials. Guided by physiological research, we then modified our model to reproduce pathological states of mild Parkinson's and Huntington's diseases. We found that in the Parkinsonian state PMC activity levels become extremely variable, which is caused by oscillations arising in the BG-thalamo-cortical loop. The model reproduced severe impairment of learning and predicted that this is caused by these oscillations as well as a reduced reward prediction signal. In the Huntington state, the potentiation of the PFC-PMC connections produced better learning, but altered BG output disrupted expression of the rewarded choices. This resulted in random switching between rewarded and unrewarded choices resembling an exploratory phase that never ended. Along with other computational studies, our results further reconcile the apparent contradiction between the critical involvement of the BG in execution of previously learned actions and yet no impairment of these actions after BG output is ablated by lesions or deep brain stimulation. We predict that the cortico-BG-thalamo-cortical loop conforms to previously learned choice in healthy conditions, but impedes those choices in disease states.
Collapse
Affiliation(s)
- Garrett Mulcahy
- Department of Mathematics, Purdue University, West Lafayette, Indiana, United States of America
| | - Brady Atwood
- Departments of Psychiatry and Pharmacology & Toxicology, IUSM, Indianapolis, Indiana, United States of America
- Indiana Alcohol Research Center, IUSM, Indianapolis, Indiana, United States of America
| | - Alexey Kuznetsov
- Indiana Alcohol Research Center, IUSM, Indianapolis, Indiana, United States of America
- Department of Mathematical Sciences, IUPUI, Indianapolis, Indiana, United States of America
| |
Collapse
|
91
|
Ryan MB, Bair-Marshall C, Nelson AB. Aberrant Striatal Activity in Parkinsonism and Levodopa-Induced Dyskinesia. Cell Rep 2019; 23:3438-3446.e5. [PMID: 29924988 DOI: 10.1016/j.celrep.2018.05.059] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 03/24/2018] [Accepted: 05/16/2018] [Indexed: 01/01/2023] Open
Abstract
Action selection relies on the coordinated activity of striatal direct and indirect pathway medium spiny neurons (dMSNs and iMSNs, respectively). Loss of dopamine in Parkinson's disease is thought to disrupt this balance. While dopamine replacement with levodopa may restore normal function, the development of involuntary movements (levodopa-induced dyskinesia [LID]) limits therapy. How chronic dopamine loss and replacement with levodopa modulate the firing of identified MSNs in behaving animals is unknown. Using optogenetically labeled striatal single-unit recordings, we assess circuit dysfunction in parkinsonism and LID. Counter to current models, we found that following dopamine depletion, iMSN firing was elevated only during periods of immobility, while dMSN firing was dramatically and persistently reduced. Most notably, we identified a subpopulation of dMSNs with abnormally high levodopa-evoked firing rates, which correlated specifically with dyskinesia. These findings provide key insights into the circuit mechanisms underlying parkinsonism and LID, with implications for developing targeted therapies.
Collapse
Affiliation(s)
- Michael B Ryan
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA
| | | | - Alexandra B Nelson
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Department of Neurology, UCSF, San Francisco, CA 94158, USA.
| |
Collapse
|
92
|
Multimodal Training Reduces Fall Frequency as Physical Activity Increases in Individuals With Parkinson's Disease. J Phys Act Health 2019; 16:1085-1091. [PMID: 31648204 DOI: 10.1123/jpah.2018-0595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 08/05/2019] [Accepted: 08/19/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Parkinson's disease (PD) results in a global decrease in information processing, ultimately resulting in dysfunction executing motor-cognitive tasks. Motor-cognitive impairments contribute to postural instability, often leading to falls and decreased physical activity. The aim of this study was to determine the effects of a multimodal training (MMT) versus single-modal (SMT) training on motor symptoms, fall frequency, and physical activity in patients with PD classified as fallers. METHODS Individuals with PD were randomized into SMT (n = 11) or MMT (n = 10) and completed training 3 times per week for 8 weeks. The SMT completed gait and cognitive training separately, whereas MMT completed gait and cognitive training simultaneously during each 45-minute session. Physical activity, 30-day fall frequency, and PD motor symptoms were assessed at baseline, posttreatment, and during a 4-week follow-up. RESULTS Both groups exhibited significant (P < .05) improvements in clinical ratings of motor function, as symptoms improved by 8% and 15% for SMT and MMT, respectively. Physical activity significantly increased (P < .05) for both groups from baseline (mean steps 4942 [4415]) to posttreatment (mean steps 5914 [5425]). The MMT resulted in a significant 60% reduction in falls. CONCLUSIONS Although SMT and MMT approaches are both effective in improving physical activity and motor symptoms of PD, only MMT reduced fall frequency after the intervention.
Collapse
|
93
|
Miterko LN, Baker KB, Beckinghausen J, Bradnam LV, Cheng MY, Cooperrider J, DeLong MR, Gornati SV, Hallett M, Heck DH, Hoebeek FE, Kouzani AZ, Kuo SH, Louis ED, Machado A, Manto M, McCambridge AB, Nitsche MA, Taib NOB, Popa T, Tanaka M, Timmann D, Steinberg GK, Wang EH, Wichmann T, Xie T, Sillitoe RV. Consensus Paper: Experimental Neurostimulation of the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2019; 18:1064-1097. [PMID: 31165428 PMCID: PMC6867990 DOI: 10.1007/s12311-019-01041-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cerebellum is best known for its role in controlling motor behaviors. However, recent work supports the view that it also influences non-motor behaviors. The contribution of the cerebellum towards different brain functions is underscored by its involvement in a diverse and increasing number of neurological and neuropsychiatric conditions including ataxia, dystonia, essential tremor, Parkinson's disease (PD), epilepsy, stroke, multiple sclerosis, autism spectrum disorders, dyslexia, attention deficit hyperactivity disorder (ADHD), and schizophrenia. Although there are no cures for these conditions, cerebellar stimulation is quickly gaining attention for symptomatic alleviation, as cerebellar circuitry has arisen as a promising target for invasive and non-invasive neuromodulation. This consensus paper brings together experts from the fields of neurophysiology, neurology, and neurosurgery to discuss recent efforts in using the cerebellum as a therapeutic intervention. We report on the most advanced techniques for manipulating cerebellar circuits in humans and animal models and define key hurdles and questions for moving forward.
Collapse
Affiliation(s)
- Lauren N Miterko
- Department of Pathology and Immunology, Department of Neuroscience, Program in Developmental Biology, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Kenneth B Baker
- Neurological Institute, Department of Neurosurgery, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Jaclyn Beckinghausen
- Department of Pathology and Immunology, Department of Neuroscience, Program in Developmental Biology, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Lynley V Bradnam
- Department of Exercise Science, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Michelle Y Cheng
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P352, Stanford, CA, 94305-5487, USA
| | - Jessica Cooperrider
- Neurological Institute, Department of Neurosurgery, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Mahlon R DeLong
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Simona V Gornati
- Department of Neuroscience, Erasmus Medical Center, 3015 AA, Rotterdam, Netherlands
| | - Mark Hallett
- Human Motor Control Section, NINDS, NIH, Building 10, Room 7D37, 10 Center Dr MSC 1428, Bethesda, MD, 20892-1428, USA
| | - Detlef H Heck
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, 855 Monroe Ave, Memphis, TN, 38163, USA
| | - Freek E Hoebeek
- Department of Neuroscience, Erasmus Medical Center, 3015 AA, Rotterdam, Netherlands
- NIDOD Department, Wilhelmina Children's Hospital, University Medical Center Utrecht Brain Center, Utrecht, Netherlands
| | - Abbas Z Kouzani
- School of Engineering, Deakin University, Geelong, VIC, 3216, Australia
| | - Sheng-Han Kuo
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Elan D Louis
- Department of Neurology, Yale School of Medicine, Department of Chronic Disease Epidemiology, Yale School of Public Health, Center for Neuroepidemiology and Clinical Research, Yale School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Andre Machado
- Neurological Institute, Department of Neurosurgery, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Mario Manto
- Service de Neurologie, CHU-Charleroi, 6000, Charleroi, Belgium
- Service des Neurosciences, Université de Mons, 7000, Mons, Belgium
| | - Alana B McCambridge
- Graduate School of Health, Physiotherapy, University of Technology Sydney, PO Box 123, Broadway, Sydney, NSW, 2007, Australia
| | - Michael A Nitsche
- Department of Psychology and Neurosiences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany
| | | | - Traian Popa
- Human Motor Control Section, NINDS, NIH, Building 10, Room 7D37, 10 Center Dr MSC 1428, Bethesda, MD, 20892-1428, USA
- Defitech Chair of Clinical Neuroengineering, Center for Neuroprosthetics (CNP) and Brain Mind Institute (BMI), Ecole Polytechnique Federale de Lausanne (EPFL), Sion, Switzerland
| | - Masaki Tanaka
- Department of Physiology, Hokkaido University School of Medicine, Sapporo, 060-8638, Japan
| | - Dagmar Timmann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P352, Stanford, CA, 94305-5487, USA
- R281 Department of Neurosurgery, Stanfod University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Eric H Wang
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P352, Stanford, CA, 94305-5487, USA
| | - Thomas Wichmann
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
| | - Tao Xie
- Department of Neurology, University of Chicago, 5841 S. Maryland Avenue, MC 2030, Chicago, IL, 60637-1470, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Department of Neuroscience, Program in Developmental Biology, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.
| |
Collapse
|
94
|
Hyland BI, Seeger-Armbruster S, Smither RA, Parr-Brownlie LC. Altered Recruitment of Motor Cortex Neuronal Activity During the Grasping Phase of Skilled Reaching in a Chronic Rat Model of Unilateral Parkinsonism. J Neurosci 2019; 39:9660-9672. [PMID: 31641050 PMCID: PMC6880456 DOI: 10.1523/jneurosci.0720-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/17/2019] [Accepted: 10/16/2019] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease causes prominent difficulties in the generation and execution of voluntary limb movements, including regulation of distal muscles and coordination of proximal and distal movement components to achieve accurate grasping. Difficulties with manual dexterity have a major impact on activities of daily living. We used extracellular single neuron recordings to investigate the neural underpinnings of parkinsonian movement deficits in the motor cortex of chronic unilateral 6-hydroxydopamine lesion male rats performing a skilled reach-to-grasp task the. Both normal movements and parkinsonian deficits in this task have striking homology to human performance. In lesioned animals there were several differences in the activity of cortical neurons during reaches by the affected limb compared with control rats. These included an increase in proportions of neurons showing rate decreases, along with increased amplitude of their average rate-decrease response at specific times during the reach, suggesting a shift in the balance of net excitation and inhibition of cortical neurons; a significant increase in the duration of rate-increase responses, which could result from reduced coupling of cortical activity to specific movement components; and changes in the timing and incidence of neurons with pure rate-increase or biphasic responses, particularly at the end of reach when grasping would normally be occurring. The changes in cortical activity may account for the deficits that occur in skilled distal motor control following dopamine depletion, and highlight the need for treatment strategies targeted toward modulating cortical mechanisms for fine distal motor control in patients.SIGNIFICANCE STATEMENT We show for the first time in a chronic lesion rat model of Parkinson's disease movement deficits that there are specific changes in motor cortex neuron activity associated with the grasping phase of a skilled motor task. Such changes provide a possible mechanism underpinning the problems with manual dexterity seen in Parkinson's patients and highlight the need for treatment strategies targeted toward distal motor control.
Collapse
Affiliation(s)
| | | | - Roseanna A Smither
- Department of Physiology and
- Department of Anatomy, School of Biomedical Science and Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand and the Brain Research New Zealand Centre of Research Excellence
| | - Louise C Parr-Brownlie
- Department of Anatomy, School of Biomedical Science and Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand and the Brain Research New Zealand Centre of Research Excellence
| |
Collapse
|
95
|
Vitanova KS, Stringer KM, Benitez DP, Brenton J, Cummings DM. Dementia associated with disorders of the basal ganglia. J Neurosci Res 2019; 97:1728-1741. [PMID: 31392765 DOI: 10.1002/jnr.24508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 01/12/2023]
Abstract
Dementia is now the leading cause of death in the United Kingdom, accounting for over 12% of all deaths and is the fifth most common cause of death worldwide. As treatments for heart disease and cancers improve and the population ages, the number of sufferers will only increase, with the chance of developing dementia doubling every 5 years after the age of 65. Finding an effective treatment is ever more critical to avert this pandemic health (and economic) crisis. To date, most dementia-related research has focused on the cortex and the hippocampus; however, with dementia becoming more fully recognized as aspects of diseases historically categorized as motor disorders (e.g., Parkinson's and Huntington's diseases), the role of the basal ganglia in dementia is coming to the fore. Conversely, it is highly likely that neuronal pathways in these structures traditionally considered as spared in Alzheimer's disease are also affected, particularly in later stages of the disease. In this review, we examine some of the limited evidence linking the basal ganglia to dementia.
Collapse
Affiliation(s)
- Karina S Vitanova
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Katie M Stringer
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Diana P Benitez
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Jonathan Brenton
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Damian M Cummings
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| |
Collapse
|
96
|
Miron-Shahar Y, Kantelhardt JW, Grinberg A, Hassin-Baer S, Blatt I, Inzelberg R, Plotnik M. Excessive phase synchronization in cortical activation during locomotion in persons with Parkinson's disease. Parkinsonism Relat Disord 2019; 65:210-216. [PMID: 31383631 DOI: 10.1016/j.parkreldis.2019.05.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is characterized by gait disturbances, which become severe during the advanced stages of the disease. Though gait impairments in Parkinson's disease have been extensively described in terms of spatiotemporal gait parameters, little is known regarding associated patterns of cortical activity. The objective of the present study is to test if interhemispheric synchronization differs between participants with PD and healthy elderly controls (NPD). We analyzed electroencephalography (EEG) signals recorded during bilateral movements, i.e., locomotion and hand tapping. METHODS Fifteen participants with PD ('OFF' their anti-parkinsonian medications) and eight NPD were assessed during quiet standing, straight-line walking, turning, and hand tapping tasks. Using a 32-electrode EEG array, we quantified the synchronization in periodic cortical activation between the brain hemispheres (interhemispheric phase synchronization; inter-PS). Theta, alpha, beta, and gamma bands were evaluated. RESULTS In all bands, inter-PS was significantly higher for the PD group as compared with the NPD group during standing and walking (p < 0.001) and during bimanual tasks (p = 0.026). CONCLUSIONS Persons with PD exhibit increased inter-PS as compared with NPD participants. These findings support previous evidence from animal studies, that bilateral cortical hypersynchronization emerges from the asymmetric neural degeneration that is at the base of the disease. Future studies should elucidate the long-term temporal development of this hypersynchronization and its clinical relevance (e.g., can it 'serve' as prodromal marker?).
Collapse
Affiliation(s)
- Yael Miron-Shahar
- School of Graduate Studies, Neuroscience Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Jan W Kantelhardt
- Institute of Physics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Adam Grinberg
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Tel Hashomer, Israel
| | - Sharon Hassin-Baer
- Movement Disorders Institute, Sagol Neuroscience Center and Department of Neurology, Sheba Medical Center, Tel-Hashomer, Israel; Department of Neurology, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ilan Blatt
- Department of Neurology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Rivka Inzelberg
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Applied Mathematics and Computer Science, the Weizmann Institute of Science, Rehovot, Israel
| | - Meir Plotnik
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Tel Hashomer, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Gonda Brain Research Center, Bar Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
97
|
Geng X, Wang X, He F, Zhang X, Xie J, Gao G, Han H, Yao X, Zhang H, Gao Y, Wang Y, Wang M. Spike and Local Field Synchronization Between the Pedunculopontine Nucleus and Primary Motor Cortex in a Rat Model of Parkinson's Disease. Neuroscience 2019; 404:470-483. [DOI: 10.1016/j.neuroscience.2019.01.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/16/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
|
98
|
Dopaminergic modulation of striatal function and Parkinson's disease. J Neural Transm (Vienna) 2019; 126:411-422. [PMID: 30937538 DOI: 10.1007/s00702-019-01997-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/20/2019] [Indexed: 01/24/2023]
Abstract
The striatum is richly innervated by mesencephalic dopaminergic neurons that modulate a diverse array of cellular and synaptic functions that control goal-directed actions and habits. The loss of this innervation has long been thought to be the principal cause of the cardinal motor symptoms of Parkinson's disease (PD). Moreover, chronic, pharmacological overstimulation of striatal dopamine (DA) receptors is generally viewed as the trigger for levodopa-induced dyskinesia (LID) in late-stage PD patients. Here, we discuss recent advances in our understanding of the relationship between the striatum and DA, particularly as it relates to PD and LID. First, it has become clear that chronic perturbations of DA levels in PD and LID bring about cell type-specific, homeostatic changes in spiny projection neurons (SPNs) that tend to normalize striatal activity. Second, perturbations in DA signaling also bring about non-homeostatic aberrations in synaptic plasticity that contribute to disease symptoms. Third, it has become evident that striatal interneurons are major determinants of network activity and behavior in PD and LID. Finally, recent work examining the activity of SPNs in freely moving animals has revealed that the pathophysiology induced by altered DA signaling is not limited to imbalance in the average spiking in direct and indirect pathways, but involves more nuanced disruptions of neuronal ensemble activity.
Collapse
|
99
|
State-Dependent Spike and Local Field Synchronization between the Thalamic Parafascicular Nucleus and the Dorsal Striatum in a Rat Model of Parkinson's Disease. Neuroscience 2019; 404:27-38. [DOI: 10.1016/j.neuroscience.2019.01.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 01/29/2023]
|
100
|
Abstract
Parkinson's disease (PD) is predominantly idiopathic in origin, and a large body of evidence indicates that gastrointestinal (GI) dysfunctions are a significant comorbid clinical feature; these dysfunctions include dysphagia, nausea, delayed gastric emptying, and severe constipation, all of which occur commonly before the onset of the well-known motor symptoms of PD. Based on a distinct distribution pattern of Lewy bodies (LB) in the enteric nervous system (ENS) and in the preganglionic neurons of the dorsal motor nucleus of the vagus (DMV), and together with the early onset of GI symptoms, it was suggested that idiopathic PD begins in the ENS and spreads to the central nervous system (CNS), reaching the DMV and the substantia nigra pars compacta (SNpc). These two areas are connected by a recently discovered monosynaptic nigro-vagal pathway, which is dysfunctional in rodent models of PD. An alternative hypothesis downplays the role of LB transport through the vagus nerve and proposes that PD pathology is governed by regional or cell-restricted factors as the leading cause of nigral neuronal degeneration. The purpose of this brief review is to summarize the neuronal electrophysiological findings in the SNpc and DMV in PD.
Collapse
Affiliation(s)
- Cecilia Bove
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine , Hershey, Pennsylvania
| | - R Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine , Hershey, Pennsylvania
| |
Collapse
|