1
|
Steiner LA, Crompton D, Sumarac S, Vetkas A, Germann J, Scherer M, Justich M, Boutet A, Popovic MR, Hodaie M, Kalia SK, Fasano A, Hutchison Wd WD, Lozano AM, Lankarany M, Kühn AA, Milosevic L. Neural signatures of indirect pathway activity during subthalamic stimulation in Parkinson's disease. Nat Commun 2024; 15:3130. [PMID: 38605039 PMCID: PMC11009243 DOI: 10.1038/s41467-024-47552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) produces an electrophysiological signature called evoked resonant neural activity (ERNA); a high-frequency oscillation that has been linked to treatment efficacy. However, the single-neuron and synaptic bases of ERNA are unsubstantiated. This study proposes that ERNA is a subcortical neuronal circuit signature of DBS-mediated engagement of the basal ganglia indirect pathway network. In people with Parkinson's disease, we: (i) showed that each peak of the ERNA waveform is associated with temporally-locked neuronal inhibition in the STN; (ii) characterized the temporal dynamics of ERNA; (iii) identified a putative mesocircuit architecture, embedded with empirically-derived synaptic dynamics, that is necessary for the emergence of ERNA in silico; (iv) localized ERNA to the dorsal STN in electrophysiological and normative anatomical space; (v) used patient-wise hotspot locations to assess spatial relevance of ERNA with respect to DBS outcome; and (vi) characterized the local fiber activation profile associated with the derived group-level ERNA hotspot.
Collapse
Affiliation(s)
- Leon A Steiner
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
- Berlin Institute of Health (BIH), Berlin, 10178, Germany
| | - David Crompton
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Srdjan Sumarac
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Artur Vetkas
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
| | - Jürgen Germann
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
| | - Maximilian Scherer
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Maria Justich
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Department of Neurology, University of Toronto, Toronto, ON, M5S 3H2, Canada
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
| | - Alexandre Boutet
- Joint Department of Medical Imaging, University of Toronto, Toronto, ON, M5G 1×6, Canada
| | - Milos R Popovic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- KITE Research Institute, University Health Network, Toronto, ON, M5G 2A2, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
| | - Mojgan Hodaie
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Suneil K Kalia
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- KITE Research Institute, University Health Network, Toronto, ON, M5G 2A2, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Alfonso Fasano
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Department of Neurology, University of Toronto, Toronto, ON, M5S 3H2, Canada
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - William D Hutchison Wd
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Andres M Lozano
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5T 2S8, Canada
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Milad Lankarany
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada
| | - Andrea A Kühn
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Luka Milosevic
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
- KITE Research Institute, University Health Network, Toronto, ON, M5G 2A2, Canada.
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5T 2S8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
2
|
Chen JL, Kuo CC. Inhibition of resurgent Na + currents by rufinamide. Neuropharmacology 2024; 247:109835. [PMID: 38228283 DOI: 10.1016/j.neuropharm.2024.109835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/18/2024]
Abstract
Na+ channels are essential for the genesis of action potentials in most neurons. After opening by membrane depolarization, Na+ channels enter a series of inactivated states (e.g. the fast, intermediate, and slow inactivated states; or If, Ii, and Is). The inactivated Na+ channel may recover via the open state upon membrane repolarization, giving rise to "resurgent" Na+ currents which could be critical for densely repetitive or burst discharges. We incubated CHO-K1 cells transfected with human NaV1.7 cDNA and measured resurgent currents with whole-cell patch recordings. We found Ii is the major inactivated state responsible for the genesis of resurgent currents. Rufinamide, in therapeutic concentrations, could selectively bind to Ii to slow the recovery process and dose-dependently inhibit resurgent currents. The other Na+ channel-inhibiting antiseizure medications (ASM), such as phenytoin and lacosamide (selectively binds to If and Is, separately), fail to show a similar inhibitory effect in clinically relevant concentrations. Resurgent currents are decreased with lengthening of the prepulse, presumably because of redistribution of the channel from Ii to If. Rufinamide could accentuate the decrease to mimic a use-dependent inhibitory effect. The molecular action of slowing of recovery from inactivation by binding to Ii also explains the highly correlative inhibitory effect of rufinamide on both transient and resurgent Na+ currents. The modest but correlative inhibition of both currents may make a novel synergistic effect and thus strong-enough suppression of pathological repetitive and especially burst discharges. Rufinamide may thus have a unique spectrum of therapeutic applications for disorders with excessive neural excitabilities.
Collapse
Affiliation(s)
- Jian-Lin Chen
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chung-Chin Kuo
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
3
|
Xu W, Wang J, Li XN, Liang J, Song L, Wu Y, Liu Z, Sun B, Li WG. Neuronal and synaptic adaptations underlying the benefits of deep brain stimulation for Parkinson's disease. Transl Neurodegener 2023; 12:55. [PMID: 38037124 PMCID: PMC10688037 DOI: 10.1186/s40035-023-00390-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
Deep brain stimulation (DBS) is a well-established and effective treatment for patients with advanced Parkinson's disease (PD), yet its underlying mechanisms remain enigmatic. Optogenetics, primarily conducted in animal models, provides a unique approach that allows cell type- and projection-specific modulation that mirrors the frequency-dependent stimulus effects of DBS. Opto-DBS research in animal models plays a pivotal role in unraveling the neuronal and synaptic adaptations that contribute to the efficacy of DBS in PD treatment. DBS-induced neuronal responses rely on a complex interplay between the distributions of presynaptic inputs, frequency-dependent synaptic depression, and the intrinsic excitability of postsynaptic neurons. This orchestration leads to conversion of firing patterns, enabling both antidromic and orthodromic modulation of neural circuits. Understanding these mechanisms is vital for decoding position- and programming-dependent effects of DBS. Furthermore, patterned stimulation is emerging as a promising strategy yielding long-lasting therapeutic benefits. Research on the neuronal and synaptic adaptations to DBS may pave the way for the development of more enduring and precise modulation patterns. Advanced technologies, such as adaptive DBS or directional electrodes, can also be integrated for circuit-specific neuromodulation. These insights hold the potential to greatly improve the effectiveness of DBS and advance PD treatment to new levels.
Collapse
Affiliation(s)
- Wenying Xu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jingxue Liang
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lu Song
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
- Ministry of Education-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
4
|
Kaur S, Sehrawat A, Mastana SS, Kandimalla R, Sharma PK, Bhatti GK, Bhatti JS. Targeting calcium homeostasis and impaired inter-organelle crosstalk as a potential therapeutic approach in Parkinson's disease. Life Sci 2023; 330:121995. [PMID: 37541578 DOI: 10.1016/j.lfs.2023.121995] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, leading to motor symptoms such as tremors, rigidity, and bradykinesia. Current therapeutic strategies for PD are limited and mainly involve symptomatic relief, with no available treatment for the underlying causes of the disease. Therefore, there is a need for new therapeutic approaches that target the underlying pathophysiological mechanisms of PD. Calcium homeostasis is an essential process for maintaining proper cellular function and survival, including neuronal cells. Calcium dysregulation is also observed in various organelles, including the endoplasmic reticulum (ER), mitochondria, and lysosomes, resulting in organelle dysfunction and impaired inter-organelle communication. The ER, as the primary calcium reservoir, is responsible for folding proteins and maintaining calcium homeostasis, and its dysregulation can lead to protein misfolding and neurodegeneration. The crosstalk between ER and mitochondrial calcium signaling is disrupted in PD, leading to neuronal dysfunction and death. In addition, a lethal network of calcium cytotoxicity utilizes mitochondria, ER and lysosome to destroy neurons. This review article focused on the complex role of calcium dysregulation and its role in aggravating functioning of organelles in PD so as to provide new insight into therapeutic strategies for treating this disease. Targeting dysfunctional organelles, such as the ER and mitochondria and lysosomes and whole network of calcium dyshomeostasis can restore proper calcium homeostasis and improve neuronal function. Additionally targeting calcium dyshomeostasis that arises from miscommunication between several organelles can be targeted so that therapeutic effects of calcium are realised in whole cellular territory.
Collapse
Affiliation(s)
- Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Abhishek Sehrawat
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, India
| | | | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
5
|
Matthews LG, Puryear CB, Correia SS, Srinivasan S, Belfort GM, Pan MK, Kuo SH. T-type calcium channels as therapeutic targets in essential tremor and Parkinson's disease. Ann Clin Transl Neurol 2023; 10:462-483. [PMID: 36738196 PMCID: PMC10109288 DOI: 10.1002/acn3.51735] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 02/05/2023] Open
Abstract
Neuronal action potential firing patterns are key components of healthy brain function. Importantly, restoring dysregulated neuronal firing patterns has the potential to be a promising strategy in the development of novel therapeutics for disorders of the central nervous system. Here, we review the pathophysiology of essential tremor and Parkinson's disease, the two most common movement disorders, with a focus on mechanisms underlying the genesis of abnormal firing patterns in the implicated neural circuits. Aberrant burst firing of neurons in the cerebello-thalamo-cortical and basal ganglia-thalamo-cortical circuits contribute to the clinical symptoms of essential tremor and Parkinson's disease, respectively, and T-type calcium channels play a key role in regulating this activity in both the disorders. Accordingly, modulating T-type calcium channel activity has received attention as a potentially promising therapeutic approach to normalize abnormal burst firing in these diseases. In this review, we explore the evidence supporting the theory that T-type calcium channel blockers can ameliorate the pathophysiologic mechanisms underlying essential tremor and Parkinson's disease, furthering the case for clinical investigation of these compounds. We conclude with key considerations for future investigational efforts, providing a critical framework for the development of much needed agents capable of targeting the dysfunctional circuitry underlying movement disorders such as essential tremor, Parkinson's disease, and beyond.
Collapse
Affiliation(s)
| | - Corey B Puryear
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA
| | | | - Sharan Srinivasan
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | - Ming-Kai Pan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, 10051, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, 10617, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, 10002, Taiwan.,Cerebellar Research Center, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, 64041, Taiwan
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, New York, 10032, USA.,Initiative for Columbia Ataxia and Tremor, Columbia University, New York, New York, 10032, USA
| |
Collapse
|
6
|
Sharma A, Rahman G, Gorelik J, Bhargava A. Voltage-Gated T-Type Calcium Channel Modulation by Kinases and Phosphatases: The Old Ones, the New Ones, and the Missing Ones. Cells 2023; 12:461. [PMID: 36766802 PMCID: PMC9913649 DOI: 10.3390/cells12030461] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/14/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Calcium (Ca2+) can regulate a wide variety of cellular fates, such as proliferation, apoptosis, and autophagy. More importantly, changes in the intracellular Ca2+ level can modulate signaling pathways that control a broad range of physiological as well as pathological cellular events, including those important to cellular excitability, cell cycle, gene-transcription, contraction, cancer progression, etc. Not only intracellular Ca2+ level but the distribution of Ca2+ in the intracellular compartments is also a highly regulated process. For this Ca2+ homeostasis, numerous Ca2+ chelating, storage, and transport mechanisms are required. There are also specialized proteins that are responsible for buffering and transport of Ca2+. T-type Ca2+ channels (TTCCs) are one of those specialized proteins which play a key role in the signal transduction of many excitable and non-excitable cell types. TTCCs are low-voltage activated channels that belong to the family of voltage-gated Ca2+ channels. Over decades, multiple kinases and phosphatases have been shown to modulate the activity of TTCCs, thus playing an indirect role in maintaining cellular physiology. In this review, we provide information on the kinase and phosphatase modulation of TTCC isoforms Cav3.1, Cav3.2, and Cav3.3, which are mostly described for roles unrelated to cellular excitability. We also describe possible potential modulations that are yet to be explored. For example, both mitogen-activated protein kinase and citron kinase show affinity for different TTCC isoforms; however, the effect of such interaction on TTCC current/kinetics has not been studied yet.
Collapse
Affiliation(s)
- Ankush Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| | - Ghazala Rahman
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| | - Julia Gorelik
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Anamika Bhargava
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| |
Collapse
|
7
|
Correa BH, Moreira CR, Hildebrand ME, Vieira LB. The Role of Voltage-Gated Calcium Channels in Basal Ganglia Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:183-201. [PMID: 35339179 PMCID: PMC10190140 DOI: 10.2174/1570159x20666220327211156] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
Calcium (Ca2+) plays a central role in regulating many cellular processes and influences cell survival. Several mechanisms can disrupt Ca2+ homeostasis to trigger cell death, including oxidative stress, mitochondrial damage, excitotoxicity, neuroinflammation, autophagy, and apoptosis. Voltage-gated Ca2+ channels (VGCCs) act as the main source of Ca2+ entry into electrically excitable cells, such as neurons, and they are also expressed in glial cells such as astrocytes and oligodendrocytes. The dysregulation of VGCC activity has been reported in both Parkinson's disease (PD) and Huntington's (HD). PD and HD are progressive neurodegenerative disorders (NDs) of the basal ganglia characterized by motor impairment as well as cognitive and psychiatric dysfunctions. This review will examine the putative role of neuronal VGCCs in the pathogenesis and treatment of central movement disorders, focusing on PD and HD. The link between basal ganglia disorders and VGCC physiology will provide a framework for understanding the neurodegenerative processes that occur in PD and HD, as well as a possible path towards identifying new therapeutic targets for the treatment of these debilitating disorders.
Collapse
Affiliation(s)
- Bernardo H.M. Correa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Roberto Moreira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
8
|
Srinivasan SR. Targeting Circuit Abnormalities in Neurodegenerative Disease. Mol Pharmacol 2023; 103:38-44. [PMID: 36310030 DOI: 10.1124/molpharm.122.000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 02/03/2023] Open
Abstract
Despite significant improvement in our ability to diagnose both common and rare neurodegenerative diseases and understand their underlying biologic mechanisms, there remains a disproportionate lack of effective treatments, reflecting the complexity of these disorders. Successfully advancing novel treatments for neurodegenerative disorders will require reconsideration of traditional approaches, which to date have focused largely on specific disease proteins or cells of origin. This article proposes reframing these diseases as conditions of dysfunctional circuitry as a complement to ongoing efforts. Specifically reviewed is how aberrant spiking is a common downstream mechanism in numerous neurodegenerative diseases, often driven by dysfunction in specific ion channels. Surgical modification of this electrical activity via deep brain stimulation is already an approved modality for many of these disorders. Therefore, restoring proper electrical activity by targeting these channels pharmacologically represents a viable strategy for intervention, not only for symptomatic management but also as a potential disease-modifying therapy. Such an approach is likely to be a promising route to treating these devastating disorders, either as monotherapy or in conjunction with current drugs. SIGNIFICANCE STATEMENT: Despite extensive research and improved understanding of the biology driving neurodegenerative disease, there has not been a concomitant increase in approved therapies. Accordingly, it is time to shift our perspective and recognize these diseases also as disorders of circuitry to further yield novel drug targets and new interventions. An approach focused on treating dysfunctional circuitry has the potential to reduce or reverse patient symptoms and potentially modify disease course.
Collapse
|
9
|
Cerebellar deep brain stimulation for movement disorders. Neurobiol Dis 2022; 175:105899. [DOI: 10.1016/j.nbd.2022.105899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
|
10
|
Nguyen NM, Duong MTH, Nguyen PL, Bui BP, Ahn HC, Cho J. Efonidipine Inhibits JNK and NF-κB Pathway to Attenuate Inflammation and Cell Migration Induced by Lipopolysaccharide in Microglial Cells. Biomol Ther (Seoul) 2022; 30:455-464. [PMID: 35993250 PMCID: PMC9424335 DOI: 10.4062/biomolther.2022.076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 11/12/2022] Open
Abstract
Efonidipine, a calcium channel blocker, is widely used for the treatment of hypertension and cardiovascular diseases. In our preliminary study using structure-based virtual screening, efonidipine was identified as a potential inhibitor of c-Jun N-terminal kinase 3 (JNK3). Although its antihypertensive effect is widely known, the role of efonidipine in the central nervous system has remained elusive. The present study investigated the effects of efonidipine on the inflammation and cell migration induced by lipopolysaccharide (LPS) using murine BV2 and human HMC3 microglial cell lines and elucidated signaling molecules mediating its effects. We found that the phosphorylations of JNK and its downstream molecule c-Jun in LPS-treated BV2 cells were declined by efonidipine, confirming the finding from virtual screening. In addition, efonidipine inhibited the LPS-induced production of pro-inflammatory factors, including interleukin-1β (IL-1β) and nitric oxide. Similarly, the IL-1β production in LPS-treated HMC3 cells was also inhibited by efonidipine. Efonidipine markedly impeded cell migration stimulated by LPS in both cells. Furthermore, it inhibited the phosphorylation of inhibitor kappa B, thereby suppressing nuclear translocation of nuclear factor-κB (NF-κB) in LPS-treated BV2 cells. Taken together, efonidipine exerts anti-inflammatory and anti-migratory effects in LPS-treated microglial cells through inhibition of the JNK/NF-κB pathway. These findings imply that efonidipine may be a potential candidate for drug repositioning, with beneficial impacts on brain disorders associated with neuroinflammation.
Collapse
Affiliation(s)
- Ngoc Minh Nguyen
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Men Thi Hoai Duong
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Phuong Linh Nguyen
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Bich Phuong Bui
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Hee-Chul Ahn
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Jungsook Cho
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| |
Collapse
|
11
|
Lai HJ, Deng CR, Wang RW, Lee LHN, Kuo CC. The genesis and functional consequences of cortico-subthalamic beta augmentation and excessive subthalamic burst discharges after dopaminergic deprivation. Exp Neurol 2022; 356:114153. [PMID: 35752209 DOI: 10.1016/j.expneurol.2022.114153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/19/2022] [Accepted: 06/18/2022] [Indexed: 11/24/2022]
Abstract
The cardinal electrophysiological signs in Parkinson's disease (PD) include augmented beta oscillations in the motor cortex-subthalamic nucleus (MC-STN) axis and excessive burst discharges in STN. We have shown that excessive STN burst discharges have a direct causal relation with the locomotor deficits in PD. To investigate the correlation between the two cardinal signs, we characterized the courses of development of the electrophysiological abnormalities in the hemiparkinsonian rat model. The loss of dopaminergic neurons develops fast, and is histologically completed within 4-7 days of the lesion. The increase in STN burst discharges is limited to the lesioned side, and follows a very similar course. In contrast, beta augmentation has a bilateral presentation, and requires 14-21 days for full development. Behaviorally, the gross locomotor deficits in open field test and limb akinesia in stepping test match the foregoing fast and slow time courses, respectively. A further look into the spike entrainment shows that the oscillations in local field potential (LFP) of the MC effectively entrain the multi-unit (MU) spikes of MC, STN and entopeduncular nucleus (EPN), a rat homolog of human globus pallidus interna (GPi), whereas the LFP of STN or EPN (GPi) cannot entrain the spikes in MC. We conclude that excessive STN burst discharges are a direct consequence, whereas beta augmentation is probably a secondary or adaptive changes in the cortico-subcortical re-entrant loops, to dopaminergic deprivation. Beta augmentation is therefore not so consistently present as excessive STN burst discharges, but could signal more delicate derangements at the level of cortical programming in PD.
Collapse
Affiliation(s)
- Hsing-Jung Lai
- Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.; Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan; National Taiwan University Hospital, Jin-Shan Branch, New Taipei, Taiwan
| | - Chuan-Rou Deng
- Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ren-Wei Wang
- Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Lan-Hsin Nancy Lee
- Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-Chin Kuo
- Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan..
| |
Collapse
|
12
|
Lee LHN, Huang CS, Wang RW, Lai HJ, Chung CC, Yang YC, Kuo CC. Deep brain stimulation rectifies the noisy cortex and irresponsive subthalamus to improve parkinsonian locomotor activities. NPJ Parkinsons Dis 2022; 8:77. [PMID: 35725730 PMCID: PMC9209473 DOI: 10.1038/s41531-022-00343-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/31/2022] [Indexed: 11/10/2022] Open
Abstract
The success of deep brain stimulation (DBS) therapy indicates that Parkinson's disease is a brain rhythm disorder. However, the manifestations of the erroneous rhythms corrected by DBS remain to be established. We found that augmentation of α rhythms and α coherence between the motor cortex (MC) and the subthalamic nucleus (STN) is characteristically prokinetic and is decreased in parkinsonian rats. In multi-unit recordings, movement is normally associated with increased changes in spatiotemporal activities rather than overall spike rates in MC. In parkinsonian rats, MC shows higher spike rates at rest but less spatiotemporal activity changes upon movement, and STN burst discharges are more prevalent, longer lasting, and less responsive to MC inputs. DBS at STN rectifies the foregoing pathological MC-STN oscillations and consequently locomotor deficits, yet overstimulation may cause behavioral restlessness. These results indicate that delicate electrophysiological considerations at both cortical and subcortical levels should be exercised for optimal DBS therapy.
Collapse
Affiliation(s)
- Lan-Hsin Nancy Lee
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Neurology, Fu Jen Catholic University Hospital, New Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ren-Wei Wang
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsing-Jung Lai
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Hospital, Jin-Shan Branch, New Taipei, Taiwan
| | - Chih-Ching Chung
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan. .,Department of Psychiatry, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan.
| | - Chung-Chin Kuo
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Lanzetti S, Di Biase V. Small Molecules as Modulators of Voltage-Gated Calcium Channels in Neurological Disorders: State of the Art and Perspectives. Molecules 2022; 27:1312. [PMID: 35209100 PMCID: PMC8879281 DOI: 10.3390/molecules27041312] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.
Collapse
Affiliation(s)
| | - Valentina Di Biase
- Institute of Pharmacology, Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Peter-Mayr Strasse 1, A-6020 Innsbruck, Austria;
| |
Collapse
|
14
|
Lee LHN, Huang CS, Chuang HH, Lai HJ, Yang CK, Yang YC, Kuo CC. An electrophysiological perspective on Parkinson's disease: symptomatic pathogenesis and therapeutic approaches. J Biomed Sci 2021; 28:85. [PMID: 34886870 PMCID: PMC8656091 DOI: 10.1186/s12929-021-00781-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD), or paralysis agitans, is a common neurodegenerative disease characterized by dopaminergic deprivation in the basal ganglia because of neuronal loss in the substantia nigra pars compacta. Clinically, PD apparently involves both hypokinetic (e.g. akinetic rigidity) and hyperkinetic (e.g. tremor/propulsion) symptoms. The symptomatic pathogenesis, however, has remained elusive. The recent success of deep brain stimulation (DBS) therapy applied to the subthalamic nucleus (STN) or the globus pallidus pars internus indicates that there are essential electrophysiological abnormalities in PD. Consistently, dopamine-deprived STN shows excessive burst discharges. This proves to be a central pathophysiological element causally linked to the locomotor deficits in PD, as maneuvers (such as DBS of different polarities) decreasing and increasing STN burst discharges would decrease and increase the locomotor deficits, respectively. STN bursts are not so autonomous but show a "relay" feature, requiring glutamatergic synaptic inputs from the motor cortex (MC) to develop. In PD, there is an increase in overall MC activities and the corticosubthalamic input is enhanced and contributory to excessive burst discharges in STN. The increase in MC activities may be relevant to the enhanced beta power in local field potentials (LFP) as well as the deranged motor programming at the cortical level in PD. Moreover, MC could not only drive erroneous STN bursts, but also be driven by STN discharges at specific LFP frequencies (~ 4 to 6 Hz) to produce coherent tremulous muscle contractions. In essence, PD may be viewed as a disorder with deranged rhythms in the cortico-subcortical re-entrant loops, manifestly including STN, the major component of the oscillating core, and MC, the origin of the final common descending motor pathways. The configurations of the deranged rhythms may play a determinant role in the symptomatic pathogenesis of PD, and provide insight into the mechanism underlying normal motor control. Therapeutic brain stimulation for PD and relevant disorders should be adaptively exercised with in-depth pathophysiological considerations for each individual patient, and aim at a final normalization of cortical discharge patterns for the best ameliorating effect on the locomotor and even non-motor symptoms.
Collapse
Affiliation(s)
- Lan-Hsin Nancy Lee
- Department of Physiology, National Taiwan University College of Medicine, 1 Jen-Ai Road, 1st Section, Taipei, 100, Taiwan.,Department of Neurology, Fu Jen Catholic University Hospital, New Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiang-Hao Chuang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsing-Jung Lai
- Department of Physiology, National Taiwan University College of Medicine, 1 Jen-Ai Road, 1st Section, Taipei, 100, Taiwan.,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Hospital, Jin-Shan Branch, New Taipei, Taiwan
| | - Cheng-Kai Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 333, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 333, Taiwan. .,Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan.
| | - Chung-Chin Kuo
- Department of Physiology, National Taiwan University College of Medicine, 1 Jen-Ai Road, 1st Section, Taipei, 100, Taiwan. .,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
15
|
Tai CH. Subthalamic burst firing: A pathophysiological target in Parkinson's disease. Neurosci Biobehav Rev 2021; 132:410-419. [PMID: 34856222 DOI: 10.1016/j.neubiorev.2021.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/16/2021] [Accepted: 11/28/2021] [Indexed: 11/27/2022]
Abstract
Understanding the pathophysiological mechanism of Parkinson's disease (PD) in the subthalamic nucleus (STN) has become a critical issue since deep brain stimulation (DBS) in this region has been proven as an effective treatment for this disease. The STN possesses a special ability to switch from the spike to the burst firing mode in response to dopamine deficiency in parkinsonism, and this STN burst is considered an electrophysiological signature of the cortico-basal ganglia circuit in the brains of PD patients. This review focuses on the role of STN burst firing in the pathophysiology of PD and during DBS. Here, we review existing literature on how STN bursts originate and the specific factors affecting their formation; how STN burst firing causes motor symptoms in PD and how interventions can rescue these symptoms. Finally, the similarities and differences between the two electrophysiological hallmarks of PD, STN burst firing and beta-oscillation, are discussed. STN burst firing should be considered as a pathophysiological target in PD during treatment with DBS.
Collapse
Affiliation(s)
- Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, No. 7, Jhongshan South Road, 100225, Taipei, Taiwan.
| |
Collapse
|
16
|
Targeting the T-type calcium channel Cav3.2 in GABAergic arcuate nucleus neurons to treat obesity. Mol Metab 2021; 54:101391. [PMID: 34767997 PMCID: PMC8640109 DOI: 10.1016/j.molmet.2021.101391] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE Cav3.2, a T-type low voltage-activated calcium channel widely expressed throughout the central nervous system, plays a vital role in neuronal excitability and various physiological functions. However, the effects of Cav3.2 on energy homeostasis remain unclear. Here, we examined the role of Cav3.2 expressed by hypothalamic GABAergic neurons in the regulation of food intake and body weight in mice and explored the underlying mechanisms. METHODS Male congenital Cana1h (the gene coding for Cav3.2) global knockout (Cav3.2KO) mice and their wild type (WT) littermates were first used for metabolic phenotyping studies. By using the CRISPR-Cas9 technique, Cav3.2 was selectively deleted from GABAergic neurons in the arcuate nucleus of the hypothalamus (ARH) by specifically overexpressing Cas9 protein and Cav3.2-targeting sgRNAs in ARH Vgat (VgatARH) neurons. These male mutants (Cav3.2KO-VgatARH) were used to determine whether Cav3.2 expressed by VgatARH neurons is required for the proper regulation of energy balance. Subsequently, we used an electrophysiological patch-clamp recording in ex vivo brain slices to explore the impact of Cav3.2KO on the cellular excitability of VgatARH neurons. RESULTS Male Cav3.2KO mice had significantly lower food intake than their WT littermate controls when fed with either a normal chow diet (NCD) or a high-fat diet (HFD). This hypophagia phenotype was associated with increased energy expenditure and decreased fat mass, lean mass, and total body weight. Selective deletion of Cav3.2 in VgatARH neurons resulted in similar feeding inhibition and lean phenotype without changing energy expenditure. These data provides an intrinsic mechanism to support the previous finding on ARH non-AgRP GABA neurons in regulating diet-induced obesity. Lastly, we found that naringenin extract, a predominant flavanone found in various fruits and herbs and known to act on Cav3.2, decreased the firing activity of VgatARH neurons and reduced food intake and body weight. These naringenin-induced inhibitions were fully blocked in Cav3.2KO-VgatARH mice. CONCLUSION Our results identified Cav3.2 expressed by VgatARH neurons as an essential intrinsic modulator for food intake and energy homeostasis, which is a potential therapeutic target in the treatment of obesity.
Collapse
|
17
|
Shao J, Liu Y, Gao D, Tu J, Yang F. Neural Burst Firing and Its Roles in Mental and Neurological Disorders. Front Cell Neurosci 2021; 15:741292. [PMID: 34646123 PMCID: PMC8502892 DOI: 10.3389/fncel.2021.741292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022] Open
Abstract
Neural firing patterns are critical for specific information coding and transmission, and abnormal firing is implicated in a series of neural pathologies. Recent studies have indicated that enhanced burst firing mediated by T-type voltage-gated calcium channels (T-VGCCs) in specific neuronal subtypes is involved in several mental or neurological disorders such as depression and epilepsy, while suppression of T-VGCCs relieve related symptoms. Burst firing consists of groups of relatively high-frequency spikes separated by quiescence. Neurons in a variety of brain areas, including the thalamus, hypothalamus, cortex, and hippocampus, display burst firing, but the ionic mechanisms that generating burst firing and the related physiological functions vary among regions. In this review, we summarize recent findings on the mechanisms underlying burst firing in various brain areas, as well as the roles of burst firing in several mental and neurological disorders. We also discuss the ion channels and receptors that may regulate burst firing directly or indirectly, with these molecules highlighted as potential intervention targets for the treatment of mental and neurological disorders.
Collapse
Affiliation(s)
- Jie Shao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Yunhui Liu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Dashuang Gao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Fan Yang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Huang CS, Wang GH, Chuang HH, Chuang AY, Yeh JY, Lai YC, Yang YC. Conveyance of cortical pacing for parkinsonian tremor-like hyperkinetic behavior by subthalamic dysrhythmia. Cell Rep 2021; 35:109007. [PMID: 33882305 DOI: 10.1016/j.celrep.2021.109007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/01/2020] [Accepted: 03/25/2021] [Indexed: 10/21/2022] Open
Abstract
Parkinson's disease is characterized by both hypokinetic and hyperkinetic symptoms. While increased subthalamic burst discharges have a direct causal relationship with the hypokinetic manifestations (e.g., rigidity and bradykinesia), the origin of the hyperkinetic symptoms (e.g., resting tremor and propulsive gait) has remained obscure. Neuronal burst discharges are presumed to be autonomous or less responsive to synaptic input, thereby interrupting the information flow. We, however, demonstrate that subthalamic burst discharges are dependent on cortical glutamatergic synaptic input, which is enhanced by A-type K+ channel inhibition. Excessive top-down-triggered subthalamic burst discharges then drive highly correlative activities bottom-up in the motor cortices and skeletal muscles. This leads to hyperkinetic behaviors such as tremors, which are effectively ameliorated by inhibition of cortico-subthalamic AMPAergic synaptic transmission. We conclude that subthalamic burst discharges play an imperative role in cortico-subcortical information relay, and they critically contribute to the pathogenesis of both hypokinetic and hyperkinetic parkinsonian symptoms.
Collapse
Affiliation(s)
- Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Guan-Hsun Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Medical Education, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
| | - Hsiang-Hao Chuang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ai-Yu Chuang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Jui-Yu Yeh
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yi-Chen Lai
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan.
| |
Collapse
|
19
|
Ortner NJ. Voltage-Gated Ca 2+ Channels in Dopaminergic Substantia Nigra Neurons: Therapeutic Targets for Neuroprotection in Parkinson's Disease? Front Synaptic Neurosci 2021; 13:636103. [PMID: 33716705 PMCID: PMC7952618 DOI: 10.3389/fnsyn.2021.636103] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
The loss of dopamine (DA)-producing neurons in the substantia nigra pars compacta (SN) underlies the core motor symptoms of the progressive movement disorder Parkinson's disease (PD). To date, no treatment to prevent or slow SN DA neurodegeneration exists; thus, the identification of the underlying factors contributing to the high vulnerability of these neurons represents the basis for the development of novel therapies. Disrupted Ca2+ homeostasis and mitochondrial dysfunction seem to be key players in the pathophysiology of PD. The autonomous pacemaker activity of SN DA neurons, in combination with low cytosolic Ca2+ buffering, leads to large somatodendritic fluctuations of intracellular Ca2+ levels that are linked to elevated mitochondrial oxidant stress. L-type voltage-gated Ca2+ channels (LTCCs) contribute to these Ca2+ oscillations in dendrites, and LTCC inhibition was beneficial in cellular and in vivo animal models of PD. However, in a recently completed phase 3 clinical trial, the dihydropyridine (DHP) LTCC inhibitor isradipine failed to slow disease progression in early PD patients, questioning the feasibility of DHPs for PD therapy. Novel evidence also suggests that R- and T-type Ca2+ channels (RTCCs and TTCCs, respectively) represent potential PD drug targets. This short review aims to (re)evaluate the therapeutic potential of LTCC, RTCC, and TTCC inhibition in light of novel preclinical and clinical data and the feasibility of available Ca2+ channel blockers to modify PD disease progression. I also summarize their cell-specific roles for SN DA neuron function and describe how their gating properties allow activity (and thus their contribution to stressful Ca2+ oscillations) during pacemaking.
Collapse
Affiliation(s)
- Nadine J. Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
Zhang W, Li SS, Han Y, Xu XH. Sex Differences in Electrophysiological Properties of Mouse Medial Preoptic Area Neurons Revealed by In Vitro Whole-cell Recordings. Neurosci Bull 2021; 37:166-182. [PMID: 32888180 PMCID: PMC7870743 DOI: 10.1007/s12264-020-00565-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/14/2020] [Indexed: 10/23/2022] Open
Abstract
Despite extensive characterization of sex differences in the medial preoptic area (mPOA) of the hypothalamus, we know surprisingly little about whether or how male and female mPOA neurons differ electrophysiologically, especially in terms of neuronal firing and behavioral pattern generation. In this study, by performing whole-cell patch clamp recordings of the mPOA, we investigated the influences of sex, cell type, and gonadal hormones on the electrophysiological properties of mPOA neurons. Notably, we uncovered significant sex differences in input resistance (male > female) and in the percentage of neurons that displayed post-inhibitory rebound (male > female). Furthermore, we found that the current mediated by the T-type Ca2+ channel (IT), which is known to underlie post-inhibitory rebound, was indeed larger in male mPOA neurons. Thus, we have identified salient electrophysiological properties of mPOA neurons, namely IT and post-inhibitory rebound, that are male-biased and likely contribute to the sexually dimorphic display of behaviors.
Collapse
Affiliation(s)
- Wen Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China
| | - Shuai-Shuai Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Han
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Hong Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China.
| |
Collapse
|
21
|
Hara N, Morino H, Matsuda Y, Satoh K, Hashimoto K, Maruyama H, Kawakami H. Zonisamide can ameliorate the voltage-dependence alteration of the T-type calcium channel Ca V3.1 caused by a mutation responsible for spinocerebellar ataxia. Mol Brain 2020; 13:163. [PMID: 33243296 PMCID: PMC7690142 DOI: 10.1186/s13041-020-00700-7] [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: 02/14/2020] [Accepted: 11/16/2020] [Indexed: 11/25/2022] Open
Abstract
Spinocerebellar ataxia (SCA) 42 is caused by a mutation in CACNA1G, which encodes the low voltage-gated calcium channel CaV3.1 (T-type). Patients with SCA42 exhibit a pure form of cerebellar ataxia. We encountered a patient with the p.Arg1715His mutation, suffering from intractable resting tremor, particularly head tremor. This symptom improved with the administration of low-dose of zonisamide (ZNS), a T-type calcium channel blocker effective for treating Parkinson’s disease and epilepsy. Previous electrophysiological studies showed that the voltage dependence of this mutant CaV3.1 was shifted toward the positive potential. This abnormal shift was considered a factor related to disease onset and symptoms. In this study, we performed whole-cell recordings of GFP-expressing HEK293T cells that expressed wild-type or mutant CaV3.1 and investigated the changes in the abnormal shift of voltage dependence of the mutant CaV3.1. The results showed that ZNS in an amount equivalent to the patient’s internal dose significantly ameliorated the abnormal shift in the mutant CaV3.1, giving values close to those in the wild-type. On the other hand, ZNS did not affect the voltage dependence of wild-type CaV3.1. Because CaV3.1 is known to be involved in tremogenesis, modulation of the voltage dependence of mutant CaV3.1 by ZNS might have contributed to improvement in the intractable tremor of our patient with SCA42. Moreover, efonidipine, another T-type calcium channel blocker, had no effect on tremors in our patient with SCA42 and did not improve the abnormal shift in the voltage dependence of the mutant CaV3.1. This indicates that ZNS is distinct from other T-type calcium channel blockers in terms of modulation of the voltage dependence of the mutant CaV3.1.
Collapse
Affiliation(s)
- Naoyuki Hara
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hiroyuki Morino
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Yukiko Matsuda
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kenichi Satoh
- The Center for Data Science Education and Research, Shiga University, 1-1-1 Banba, Hikone, Shiga, 522-8522, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Hideshi Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| |
Collapse
|
22
|
Hyperpolarization of the subthalamic nucleus alleviates hyperkinetic movement disorders. Sci Rep 2020; 10:8278. [PMID: 32427942 PMCID: PMC7237462 DOI: 10.1038/s41598-020-65211-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/20/2020] [Indexed: 11/08/2022] Open
Abstract
Modulation of subthalamic nucleus (STN) firing patterns with injections of depolarizing currents into the STN is an important advance for the treatment of hypokinetic movement disorders, especially Parkinson’s disease (PD). Chorea, ballism and dystonia are prototypical examples of hyperkinetic movement disorders. In our previous study, normal rats without nigro-striatal lesion were rendered hypokinetic with hyperpolarizing currents injected into the STN. Therefore, modulation of the firing pattern by injection of a hyperpolarizing current into the STN could be an effective treatment for hyperkinetic movement disorders. We investigated the effect of injecting a hyperpolarizing current into the STNs of two different types of hyperkinetic animal models and a patient with an otherwise uncontrollable hyperkinetic disorder. The two animal models included levodopa-induced hyperkinetic movement in parkinsonian rats (L-DOPA-induced dyskinesia model) and hyperkinesia induced by an intrastriatal injection of 3-nitropropionic acid (Huntington disease model), covering neurodegeneration-related as well as neurotoxin-induced derangement in the cortico-subcortical re-entrant loops. Delivering hyperpolarizing currents into the STN readily alleviated the hyperkinetic behaviors in the two animal models and in the clinical case, with an evident increase in subthalamic burst discharges in electrophysiological recordings. Application of a hyperpolarizing current into the STN via a Deep brain stimulation (DBS) electrode could be an effective general therapy for a wide spectrum of hyperkinetic movement disorders.
Collapse
|
23
|
Miranda AS, Cardozo PL, Silva FR, de Souza JM, Olmo IG, Cruz JS, Gomez MV, Ribeiro FM, Vieira LB. Alterations of Calcium Channels in a Mouse Model of Huntington's Disease and Neuroprotection by Blockage of Ca V1 Channels. ASN Neuro 2020; 11:1759091419856811. [PMID: 31216184 PMCID: PMC6585245 DOI: 10.1177/1759091419856811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative autosomal dominant disorder, characterized by symptoms of involuntary movement of the body, loss of cognitive function, psychiatric disorder, leading inevitably to death. It has been previously described that higher levels of brain expression of Cav1 channels are involved in major neurodegenerative disorders, such as Alzheimer’s disease and Parkinson’s disease. Our results demonstrate that a bacterial artificial chromosome (BAC)-mediated transgenic mouse model (BACHD mice) at the age of 3 and 12 months exhibits significantly increased Cav1.2 protein levels in the cortex, as compared with wild-type littermates. Importantly, electrophysiological analyses confirm a significant increase in L-type Ca2+ currents and total Ca2+ current density in cortical neurons from BACHD mice. By using an in vitro assay to measure neuronal cell death, we were able to observe neuronal protection against glutamate toxicity after treatment with Cav1 blockers, in wild-type and, more importantly, in BACHD neurons. According to our data, Cav1 blockers may offer an interesting strategy for the treatment of HD. Altogether, our results show that mutant huntingtin (mHtt) expression may cause a dysregulation of Cav1.2 channels and we hypothesize that this contributes to neurodegeneration during HD.
Collapse
Affiliation(s)
- Artur S Miranda
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo Leal Cardozo
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flavia R Silva
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jessica M de Souza
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella G Olmo
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jader S Cruz
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Fabiola M Ribeiro
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciene B Vieira
- 3 Department of Pharmacology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
24
|
Robles-Gómez AA, Vega AV, Florán B, Barral J. Differential calcium channel-mediated dopaminergic modulation in the subthalamonigral synapse. Synapse 2020; 74:e22149. [PMID: 31975491 DOI: 10.1002/syn.22149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/09/2019] [Accepted: 01/18/2020] [Indexed: 11/09/2022]
Abstract
Dopamine (DA) modulates basal ganglia (BG) activity for initiation and execution of goal-directed movements and habits. While most studies are aimed to striatal function, the cellular and molecular mechanisms underlying dopaminergic regulation in other nuclei of the BG are not well understood. Therefore, we set to analyze the dopaminergic modulation occurring in subthalamo-nigral synapse, in both pars compacta (SNc) and pars reticulata (SNr) neurons, because these synapses are important for the integration of information previously processed in striatum and globus pallidus. In this study, electrophysiological and pharmacological evidence of dopaminergic modulation on glutamate release through calcium channels is presented. Using paired pulse ratio (PPR) measurements and selective blockers of these ionic channels, together with agonists and antagonists of DA D2 -like receptors, we found that blockade of the CaV 3 family occludes the presynaptic inhibition produced by the activation of DA receptors pharmacologically profiled as D3 -type in the STh-SNc synapses. On the contrast, the blockade of CaV 2 channels, but not CaV 3, occlude with the effect of the D3 agonist, PD 128907, in the STh-SNr synapse. The functional role of this differential distribution of calcium channels that modulate the release of glutamate in the SN implies a fine adjustment of firing for both classes of neurons. Dopaminergic neurons of the SNc establish a DA tone within the SN based on the excitatory/inhibitory inputs; such tone may contribute to processing information from subthalamic nucleus and could also be involved in pathological DA depletion that drives hyperexcitation of SNr neurons.
Collapse
Affiliation(s)
| | - Ana V Vega
- Carrera de Médico Cirujano, FES Iztacala, UNAM, Mexico City, Mexico
| | - Benjamín Florán
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | - Jaime Barral
- Neurociencias, FES Iztacala, UNAM, Tlalnepantla de Baz, Mexico
| |
Collapse
|
25
|
Leandrou E, Emmanouilidou E, Vekrellis K. Voltage-Gated Calcium Channels and α-Synuclein: Implications in Parkinson's Disease. Front Mol Neurosci 2019; 12:237. [PMID: 31649506 PMCID: PMC6794345 DOI: 10.3389/fnmol.2019.00237] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (α-syn) is biochemically and genetically linked to Parkinson's disease (PD) and other synucleinopathies. It is now widely accepted that α-syn can be released in the extracellular space, even though the mechanism of its release is still unclear. In addition, pathology-related aggregated species of α-syn have been shown to propagate between neurons in synaptically connected areas of the brain thereby assisting the spreading of pathology in healthy neighboring neuronal cells. In neurons, calcium channels are key signaling elements that modulate the release of bioactive molecules (hormones, proteins, and neurotransmitters) through calcium sensing. Such calcium sensing activity is determined by the distinct biophysical and pharmacological properties and the ability of calcium channels to interact with other modulatory proteins. Although the function of extracellular α-syn is currently unknown, previous work suggested the presence of a calcium-dependent mechanism for α-syn secretion both in vitro, in neuronal cells in culture, and also in vivo, in the context of a trans-neuronal network in brain. Mechanisms regulating extracellular α-syn levels may be of particular importance as they could represent novel therapeutic targets. We discuss here how calcium channel activity may contribute to α-syn aggregation and secretion as a pathway to disease progression in synucleinopathies.
Collapse
Affiliation(s)
- Emmanouela Leandrou
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Evangelia Emmanouilidou
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece.,Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Vekrellis
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| |
Collapse
|
26
|
Milosevic L, Kalia SK, Hodaie M, Lozano A, Popovic MR, Hutchison W. Subthalamic suppression defines therapeutic threshold of deep brain stimulation in Parkinson's disease. J Neurol Neurosurg Psychiatry 2019; 90:1105-1108. [PMID: 31422369 DOI: 10.1136/jnnp-2019-321140] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Subthalamic deep brain stimulation (DBS) is beneficial when delivered at a high frequency. However, the effects of current amplitude and pulse width on subthalamic neuronal activity during high-frequency stimulation have not been investigated. METHODS In 20 patients with Parkinson's disease each undergoing subthalamic DBS, we recorded single-unit subthalamic activity using one microelectrode, while a separate microelectrode was used to deliver 5-10 s trains of stimulation at 100 Hz using varying current amplitudes and pulse widths (44 neurons investigated). RESULTS Analysis of variance tests confirmed significant (p<0.001) main effects of both current amplitude and pulse width on subthalamic neuronal firing during stimulation and on poststimulus inhibitory silent periods. Prolonged silent periods were often followed by postinhibitory rebound burst excitations. Additionally, a significant (p<0.0001) correlation was found between neuronal firing and total electrical energy delivered (TEED). With TEED values≤31.2 µJ/s (associated with DBS parameters of ≤2.0 mA, 130 Hz stimulation frequency and 60 µs pulse width, assuming 1 kΩ impedance), neuronal firing was sustained at a rate of 32.4%±3.3% (mean±SE), while with values>31.2 µJ/s, neurons fired at only 4.3%±1.2%. CONCLUSIONS Neuronal suppression is likely an important mechanism of action of therapeutically beneficial subthalamic DBS, which may underlie clinically relevant behavioural changes.
Collapse
Affiliation(s)
- Luka Milosevic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Suneil K Kalia
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - Mojgan Hodaie
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - Andres Lozano
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - Milos R Popovic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - William Hutchison
- Krembil Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Papapetropoulos S, Lee MS, Boyer S, Newbold EJ. A Phase 2, Randomized, Double-Blind, Placebo-Controlled Trial of CX-8998, a Selective Modulator of the T-Type Calcium Channel in Inadequately Treated Moderate to Severe Essential Tremor: T-CALM Study Design and Methodology for Efficacy Endpoint and Digital Biomarker Selection. Front Neurol 2019; 10:597. [PMID: 31244760 PMCID: PMC6579833 DOI: 10.3389/fneur.2019.00597] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/21/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Essential tremor (ET) is a common, progressive neurological syndrome with bilateral upper-limb dysfunction of at least 3-year duration, with or without tremor in other body locations. This disorder has a negative impact on daily function and quality of life. A single oral therapy has been approved by FDA for ET. Off-label pharmacotherapies have inadequate efficacy and poor tolerability with high rates of patient dissatisfaction and discontinuation. Safe and efficacious pharmacotherapies are urgently needed to decrease tremor and improve daily living. T-CALM (Tremor-CAv3 modulation) protocol is designed to assess safety and efficacy of CX-8998, a selective modulator of the T-type calcium channel, for ET therapy. Methods/Design: T-CALM is a phase 2, proof of concept, randomized, double-blind, placebo-controlled trial. Titrated doses of CX-8998 to 10 mg BID or placebo will be administered for 28 days to moderate to severe ET patients who are inadequately treated with existing therapies. The primary endpoint will be change from baseline to day 28 of The Essential Tremor Rating Assessment Performance Subscale (TETRAS-PS). Secondary efficacy endpoints for clinician and patient perception of daily function will include TETRAS Activity of Daily Living (ADL), Quality of Life in Essential Tremor Questionnaire (QUEST), Clinical Global Impression-Improvement (CGI-I), Patient Global Impression of Change (PGIC), and Goal Attainment Scale (GAS). Kinesia One, Kinesia 360, and iMotor will biometrically evaluate motor function and tremor amplitude. Safety will be assessed by adverse events, physical and neurological exams and laboratory tests. Sample size of 43 patients per group is estimated to have 90% power to detect a 5.5-point difference between CX-8998 and placebo for TETRAS-PS. Efficacy analyses will be performed with covariance (ANCOVA) and 2-sided test at 0.05 significance level. Discussion: T-CALM has a unique design with physician rating scales, patient-focused questionnaires and scales and objective motor measurements to assess clinically meaningful and congruent efficacy. Patient perception of ET debilitation and therapy with CX-8998 will be key findings. Overall goal of T-CALM is generation of safety and efficacy data to support a go, no-go decision to further develop CX-8998 for ET. Design of T-CALM may guide future clinical studies of ET pharmacotherapies. Clinical Trial Registration:www.ClinicalTrials.gov, identifier: NCT03101241
Collapse
|
28
|
Tai CH, Bellesi M, Chen AC, Lin CL, Li HH, Lin PJ, Liao WC, Hung CS, Schwarting RK, Ho YJ. A new avenue for treating neuronal diseases: Ceftriaxone, an old antibiotic demonstrating behavioral neuronal effects. Behav Brain Res 2019; 364:149-156. [DOI: 10.1016/j.bbr.2019.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/14/2019] [Accepted: 02/12/2019] [Indexed: 12/27/2022]
|
29
|
Zhuang QX, Li GY, Li B, Zhang CZ, Zhang XY, Xi K, Li HZ, Wang JJ, Zhu JN. Regularizing firing patterns of rat subthalamic neurons ameliorates parkinsonian motor deficits. J Clin Invest 2018; 128:5413-5427. [PMID: 30226827 DOI: 10.1172/jci99986] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022] Open
Abstract
The subthalamic nucleus (STN) is an effective therapeutic target for deep brain stimulation (DBS) for Parkinson's disease (PD), and histamine levels are elevated in the basal ganglia in PD patients. However, the effect of endogenous histaminergic modulation on STN neuronal activities and the neuronal mechanism underlying STN-DBS are unknown. Here, we report that STN neuronal firing patterns are more crucial than firing rates for motor control. Histamine excited STN neurons, but paradoxically ameliorated parkinsonian motor deficits, which we attributed to regularizing firing patterns of STN neurons via the hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) channel coupled to the H2 receptor. Intriguingly, DBS increased histamine release in the STN and regularized STN neuronal firing patterns under parkinsonian conditions. HCN2 contributed to the DBS-induced regularization of neuronal firing patterns, suppression of excessive β oscillations, and alleviation of motor deficits in PD. The results reveal an indispensable role for regularizing STN neuronal firing patterns in amelioration of parkinsonian motor dysfunction and a functional compensation for histamine in parkinsonian basal ganglia circuitry. The findings provide insights into mechanisms of STN-DBS as well as potential therapeutic targets and STN-DBS strategies for PD.
Collapse
Affiliation(s)
- Qian-Xing Zhuang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Guang-Ying Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Bin Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Chang-Zheng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Kang Xi
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, and.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Cáceres-Chávez VA, Hernández-Martínez R, Pérez-Ortega J, Herrera-Valdez MA, Aceves JJ, Galarraga E, Bargas J. Acute dopamine receptor blockade in substantia nigra pars reticulata: a possible model for drug-induced Parkinsonism. J Neurophysiol 2018; 120:2922-2938. [PMID: 30256736 DOI: 10.1152/jn.00579.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dopamine (DA) depletion modifies the firing pattern of neurons in the substantia nigra pars reticulata (SNr), shifting their mostly tonic firing toward irregularity and bursting, traits of pathological firing underlying rigidity and postural instability in Parkinson's disease (PD) patients and animal models of Parkinsonism (PS). Drug-induced Parkinsonism (DIP) represents 20-40% of clinical cases of PS, becoming a problem for differential diagnosis, and is still not well studied with physiological tools. It may co-occur with tardive dyskinesia. Here we use in vitro slice preparations including the SNr to observe drug-induced pathological firing by using drugs that most likely produce it, DA-receptor antagonists (SCH23390 plus sulpiride), to compare with firing patterns found in DA-depleted tissue. The hypothesis is that SNr firing would be similar under both conditions, a prerequisite to the proposal of a similar preparation to test other DIP-producing drugs. Firing was analyzed with three complementary metrics, showing similarities between DA depletion and acute DA-receptor blockade. Moreover, blockade of either nonselective cationic channels or Cav3 T-type calcium channels hyperpolarized the membrane and abolished bursting and irregular firing, silencing SNr neurons in both conditions. Therefore, currents generating firing in control conditions are in part responsible for pathological firing. Haloperidol, a DIP-producing drug, reproduced DA-receptor antagonist firing modifications. Since acute DA-receptor blockade induces SNr neuron firing similar to that found in the 6-hydroxydopamine model of PS, output basal ganglia neurons may play a role in generating DIP. Therefore, this study opens the way to test other DIP-producing drugs. NEW & NOTEWORTHY Dopamine (DA) depletion enhances substantia nigra pars reticulata (SNr) neuron bursting and irregular firing, hallmarks of Parkinsonism. Several drugs, including antipsychotics, antidepressants, and calcium channel antagonists, among others, produce drug-induced Parkinsonism. Here we show the first comparison between SNr neuron firing after DA depletion vs. firing found after acute blockade of DA receptors. It was found that firing in both conditions is similar, implying that pathological SNr neuron firing is also a physiological correlate of drug-induced Parkinsonism.
Collapse
Affiliation(s)
| | - Ricardo Hernández-Martínez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| | - Jesús Pérez-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México , Querétaro, México
| | - Marco Arieli Herrera-Valdez
- Departamento de Matemáticas, Facultad de Ciencias, Universidad Nacional Autónoma de México , México City, México
| | - Jose J Aceves
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , Mexico City, México
| | - Elvira Galarraga
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| | - José Bargas
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| |
Collapse
|
31
|
Drion G, Dethier J, Franci A, Sepulchre R. Switchable slow cellular conductances determine robustness and tunability of network states. PLoS Comput Biol 2018; 14:e1006125. [PMID: 29684009 PMCID: PMC5940245 DOI: 10.1371/journal.pcbi.1006125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 05/08/2018] [Accepted: 04/06/2018] [Indexed: 11/21/2022] Open
Abstract
Neuronal information processing is regulated by fast and localized fluctuations of brain states. Brain states reliably switch between distinct spatiotemporal signatures at a network scale even though they are composed of heterogeneous and variable rhythms at a cellular scale. We investigated the mechanisms of this network control in a conductance-based population model that reliably switches between active and oscillatory mean-fields. Robust control of the mean-field properties relies critically on a switchable negative intrinsic conductance at the cellular level. This conductance endows circuits with a shared cellular positive feedback that can switch population rhythms on and off at a cellular resolution. The switch is largely independent from other intrinsic neuronal properties, network size and synaptic connectivity. It is therefore compatible with the temporal variability and spatial heterogeneity induced by slower regulatory functions such as neuromodulation, synaptic plasticity and homeostasis. Strikingly, the required cellular mechanism is available in all cell types that possess T-type calcium channels but unavailable in computational models that neglect the slow kinetics of their activation. Brain information processing involves electrophysiological signals at multiple temporal and spatial timescales, from the single neuron level to whole brain areas. A fast and local control of these signals by neurochemicals called neuromodulators is essential in complex tasks such as movement initiation and attentional focus. The neuromodulators act at the cellular scale to control signals that propagate at potentially much larger scales. The present paper highlights the critical role of a cellular switch of excitability for the fast and localized control of cellular and network states. By turning ON and OFF the cellular switch, neuromodulators can robustly switch large populations between distinct network states. We stress the importance of controlling the switch at a cellular level and independently of the connectivity to allow for tunable spatiotemporal signatures of the network states.
Collapse
Affiliation(s)
- Guillaume Drion
- Department of Electrical Engineering and Computer Science, University of Liege, Liege, Belgium
| | - Julie Dethier
- Department of Electrical Engineering and Computer Science, University of Liege, Liege, Belgium
| | - Alessio Franci
- National Autonomous University of Mexico, Science Faculty, Department of Mathematics, Coyoacán, D.F., México
| | - Rodolphe Sepulchre
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
33
|
Schampel A, Kuerten S. Danger: High Voltage-The Role of Voltage-Gated Calcium Channels in Central Nervous System Pathology. Cells 2017; 6:E43. [PMID: 29140302 PMCID: PMC5755501 DOI: 10.3390/cells6040043] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely distributed within the central nervous system (CNS) and presumed to play an important role in the pathophysiology of a broad spectrum of CNS disorders including Alzheimer's and Parkinson's disease as well as multiple sclerosis. Several calcium channel blockers have been in clinical practice for many years so that their toxicity and side effects are well studied. However, these drugs are primarily used for the treatment of cardiovascular diseases and most if not all effects on brain functions are secondary to peripheral effects on blood pressure and circulation. While the use of calcium channel antagonists for the treatment of CNS diseases therefore still heavily depends on the development of novel strategies to specifically target different channels and channel subunits, this review is meant to provide an impulse to further emphasize the importance of future research towards this goal.
Collapse
Affiliation(s)
- Andrea Schampel
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg 97070, Germany.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91054, Germany.
| |
Collapse
|
34
|
Kim J, Kim Y, Nakajima R, Shin A, Jeong M, Park AH, Jeong Y, Jo S, Yang S, Park H, Cho SH, Cho KH, Shim I, Chung JH, Paik SB, Augustine GJ, Kim D. Inhibitory Basal Ganglia Inputs Induce Excitatory Motor Signals in the Thalamus. Neuron 2017; 95:1181-1196.e8. [PMID: 28858620 DOI: 10.1016/j.neuron.2017.08.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/07/2017] [Accepted: 08/15/2017] [Indexed: 10/19/2022]
Abstract
Basal ganglia (BG) circuits orchestrate complex motor behaviors predominantly via inhibitory synaptic outputs. Although these inhibitory BG outputs are known to reduce the excitability of postsynaptic target neurons, precisely how this change impairs motor performance remains poorly understood. Here, we show that optogenetic photostimulation of inhibitory BG inputs from the globus pallidus induces a surge of action potentials in the ventrolateral thalamic (VL) neurons and muscle contractions during the post-inhibitory period. Reduction of the neuronal population with this post-inhibitory rebound firing by knockout of T-type Ca2+ channels or photoinhibition abolishes multiple motor responses induced by the inhibitory BG input. In a low dopamine state, the number of VL neurons showing post-inhibitory firing increases, while reducing the number of active VL neurons via photoinhibition of BG input, effectively prevents Parkinson disease (PD)-like motor symptoms. Thus, BG inhibitory input generates excitatory motor signals in the thalamus and, in excess, promotes PD-like motor abnormalities. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Jeongjin Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; Center for Neuroscience, KIST, Seoul 02792, Republic of Korea
| | - Youngsoo Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Ryuichi Nakajima
- Center for Functional Connectomics, KIST, Seoul 02792, Republic of Korea
| | - Anna Shin
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Minju Jeong
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Ah Hyung Park
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Yongcheol Jeong
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Seonmi Jo
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Seungkyoung Yang
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Hosung Park
- School of Computing, KAIST, Daejeon 34141, Republic of Korea
| | - Sung-Hwan Cho
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Insop Shim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Jae Hoon Chung
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - George J Augustine
- Center for Functional Connectomics, KIST, Seoul 02792, Republic of Korea; Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore; Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Daesoo Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
35
|
Huang CS, Wang GH, Tai CH, Hu CC, Yang YC. Antiarrhythmics cure brain arrhythmia: The imperativeness of subthalamic ERG K + channels in parkinsonian discharges. SCIENCE ADVANCES 2017; 3:e1602272. [PMID: 28508055 PMCID: PMC5425237 DOI: 10.1126/sciadv.1602272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 03/15/2017] [Indexed: 06/07/2023]
Abstract
ERG K+ channels have long been known to play a crucial role in shaping cardiac action potentials and, thus, appropriate heart rhythms. The functional role of ERG channels in the central nervous system, however, remains elusive. We demonstrated that ERG channels exist in subthalamic neurons and have similar gating characteristics to those in the heart. ERG channels contribute crucially not only to the setting of membrane potential and, consequently, the firing modes, but also to the configuration of burst discharges and, consequently, the firing frequency and automaticity of the subthalamic neurons. Moreover, modulation of subthalamic discharges via ERG channels effectively modulates locomotor behaviors. ERG channel inhibitors ameliorate parkinsonian symptoms, whereas enhancers render normal animals hypokinetic. Thus, ERG K+ channels could be vital to the regulation of both cardiac and neuronal rhythms and may constitute an important pathophysiological basis and pharmacotherapeutic target for the growing list of neurological disorders related to "brain arrhythmias."
Collapse
Affiliation(s)
- Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Guan-Hsun Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Chang Hu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| |
Collapse
|
36
|
Galvan A, Devergnas A, Pittard D, Masilamoni G, Vuong J, Daniels JS, Morrison RD, Lindsley CW, Wichmann T. Lack of Antiparkinsonian Effects of Systemic Injections of the Specific T-Type Calcium Channel Blocker ML218 in MPTP-Treated Monkeys. ACS Chem Neurosci 2016; 7:1543-1551. [PMID: 27596273 DOI: 10.1021/acschemneuro.6b00186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Dopaminergic medications ameliorate many of the motor impairments of Parkinson's disease (PD). However, parkinsonism is often only partially reversed by these drugs, and they can have significant side effects. Therefore, a need remains for novel treatments of parkinsonism. Studies in rodents and preliminary clinical evidence have shown that T-type calcium channel (TTCC) antagonists have antiparkinsonian effects. However, most of the available studies utilized nonselective agents. We now evaluated whether systemic injections of the specific TTCC blocker ML218 have antiparkinsonian effects in MPTP-treated parkinsonian Rhesus monkeys. The animals were treated chronically with MPTP until they reached stable parkinsonism. In pharmacokinetic studies, we found that ML218 reaches a peak CSF concentration 1-2 h after s.c. administration. In electrocardiographic studies, we found no effects of ML218 on cardiac rhythmicity. As expected, systemic injections of the dopamine precursor L-DOPA dose-dependently increased the movements in our parkinsonian animals. We then tested the behavioral effects of systemic injections of ML218 (1, 10, or 30 mg/kg) or its vehicle, but did not detect specific antiparkinsonian effects. ML218 (3 or 10 mg/kg) was also not synergistic with L-DOPA. Using recordings of electrocorticogram signals (in one animal), we found that ML218 increased sleep. We conclude that ML218 does not have antiparkinsonian effects in MPTP-treated parkinsonian monkeys, due at least in part, to the agent's sedative effects.
Collapse
Affiliation(s)
| | | | | | | | | | - J. Scott Daniels
- Department
of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Ryan D. Morrison
- Department
of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Department
of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | | |
Collapse
|
37
|
Rangel-Barajas C, Estrada-Sánchez AM, Barton SJ, Luedtke RR, Rebec GV. Dysregulated corticostriatal activity in open-field behavior and the head-twitch response induced by the hallucinogen 2,5-dimethoxy-4-iodoamphetamine. Neuropharmacology 2016; 113:502-510. [PMID: 27816502 DOI: 10.1016/j.neuropharm.2016.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/19/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022]
Abstract
The substituted amphetamine, 2,5-dimethoxy-4-iodoamphetamine (DOI), is a hallucinogen that has been used to model a variety of psychiatric conditions. Here, we studied the effect of DOI on neural activity recorded simultaneously in the primary motor cortex (M1) and dorsal striatum of freely behaving FvB/N mice. DOI significantly decreased the firing rate of individually isolated neurons in M1 and dorsal striatum relative to pre-drug baseline. It also induced a bursting pattern of activity by increasing both the number of spikes within a burst and burst duration. In addition, DOI increased coincident firing between simultaneously recorded neuron pairs within the striatum and between M1 and dorsal striatum. Local field potential (LFP) activity also increased in coherence between M1 and dorsal striatum after DOI in the low frequency gamma band (30-50 Hz), while corticostriatal coherence in delta, theta, alpha, and beta activity decreased. We also assessed corticostriatal LFP activity in relation to the DOI-induced head-twitch response (HTR), a readily identifiable behavior used to assess potential treatments for the conditions it models. The HTR was associated with increased delta and decreased theta power in both M1 and dorsal striatum. Together, our results suggest that DOI dysregulates corticostriatal communication and that the HTR is associated with this dysregulation.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Indiana University Bloomington, Department of Psychological and Brain Sciences, Program in Neuroscience, 1101 E. 10th St., Bloomington, IN 47405, USA
| | - Ana María Estrada-Sánchez
- Indiana University Bloomington, Department of Psychological and Brain Sciences, Program in Neuroscience, 1101 E. 10th St., Bloomington, IN 47405, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Scott J Barton
- Indiana University Bloomington, Department of Psychological and Brain Sciences, Program in Neuroscience, 1101 E. 10th St., Bloomington, IN 47405, USA
| | - Robert R Luedtke
- University of North Texas Health Science Center, The Department of Pharmacology and Neuroscience, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - George V Rebec
- Indiana University Bloomington, Department of Psychological and Brain Sciences, Program in Neuroscience, 1101 E. 10th St., Bloomington, IN 47405, USA.
| |
Collapse
|
38
|
Pan MK, Kuo SH, Tai CH, Liou JY, Pei JC, Chang CY, Wang YM, Liu WC, Wang TR, Lai WS, Kuo CC. Neuronal firing patterns outweigh circuitry oscillations in parkinsonian motor control. J Clin Invest 2016; 126:4516-4526. [PMID: 27797341 DOI: 10.1172/jci88170] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/22/2016] [Indexed: 11/17/2022] Open
Abstract
Neuronal oscillations at beta frequencies (20-50 Hz) in the cortico-basal ganglia circuits have long been the leading theory for bradykinesia, the slow movements that are cardinal symptoms in Parkinson's disease (PD). The beta oscillation theory helped to drive a frequency-based design in the development of deep brain stimulation therapy for PD. However, in contrast to this theory, here we have found that bradykinesia can be completely dissociated from beta oscillations in rodent models. Instead, we observed that bradykinesia is causatively regulated by the burst-firing pattern of the subthalamic nucleus (STN) in a feed-forward, or efferent-only, mechanism. Furthermore, STN burst-firing and beta oscillations are two independent mechanisms that are regulated by different NMDA receptors in STN. Our results shift the understanding of bradykinesia pathophysiology from an interactive oscillatory theory toward a feed-forward mechanism that is coded by firing patterns. This distinct mechanism may improve understanding of the fundamental concepts of motor control and enable more selective targeting of bradykinesia-specific mechanisms to improve PD therapy.
Collapse
|
39
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
40
|
Devergnas A, Chen E, Ma Y, Hamada I, Pittard D, Kammermeier S, Mullin AP, Faundez V, Lindsley CW, Jones C, Smith Y, Wichmann T. Anatomical localization of Cav3.1 calcium channels and electrophysiological effects of T-type calcium channel blockade in the motor thalamus of MPTP-treated monkeys. J Neurophysiol 2015; 115:470-85. [PMID: 26538609 DOI: 10.1152/jn.00858.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022] Open
Abstract
Conventional anti-Parkinsonian dopamine replacement therapy is often complicated by side effects that limit the use of these medications. There is a continuing need to develop nondopaminergic approaches to treat Parkinsonism. One such approach is to use medications that normalize dopamine depletion-related firing abnormalities in the basal ganglia-thalamocortical circuitry. In this study, we assessed the potential of a specific T-type calcium channel blocker (ML218) to eliminate pathologic burst patterns of firing in the basal ganglia-receiving territory of the motor thalamus in Parkinsonian monkeys. We also carried out an anatomical study, demonstrating that the immunoreactivity for T-type calcium channels is strongly expressed in the motor thalamus in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys. At the electron microscopic level, dendrites accounted for >90% of all tissue elements that were immunoreactive for voltage-gated calcium channel, type 3.2-containing T-type calcium channels in normal and Parkinsonian monkeys. Subsequent in vivo electrophysiologic studies in awake MPTP-treated Parkinsonian monkeys demonstrated that intrathalamic microinjections of ML218 (0.5 μl of a 2.5-mM solution, injected at 0.1-0.2 μl/min) partially normalized the thalamic activity by reducing the proportion of rebound bursts and increasing the proportion of spikes in non-rebound bursts. The drug also attenuated oscillatory activity in the 3-13-Hz frequency range and increased gamma frequency oscillations. However, ML218 did not normalize Parkinsonism-related changes in firing rates and oscillatory activity in the beta frequency range. Whereas the described changes are promising, a more complete assessment of the cellular and behavioral effects of ML218 (or similar drugs) is needed for a full appraisal of their anti-Parkinsonian potential.
Collapse
Affiliation(s)
- Annaelle Devergnas
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia;
| | - Erdong Chen
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Yuxian Ma
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Ikuma Hamada
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Damien Pittard
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia
| | - Stefan Kammermeier
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Klinikum der Universität München, Neurologische Klinik und Poliklinik, München, Germany
| | - Ariana P Mullin
- Department of Cell Biology, Emory University, Atlanta, Georgia
| | - Victor Faundez
- Department of Cell Biology, Emory University, Atlanta, Georgia; Center for Social Translational Neuroscience, Emory University, Atlanta, Georgia
| | - Craig W Lindsley
- Department of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Carrie Jones
- Department of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Atlanta, Georgia; Udall Center of Excellence for Parkinson's Disease Research at Emory University, Atlanta, Georgia; Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
41
|
Murata M, Hasegawa K, Kanazawa I, Shirakura K, Kochi K, Shimazu R, Kimura T, Yoshida K, Abe T, Kurita K, Yoshizawa K, Tamaoka A, Nakano I, Shimizu T, Hattori N, Mizusawa H, Kuno S, Yokochi F, Hirabayashi K, Horiuchi E, Kawashima N, Koike R, Ishikawa A, Kuriyama M, Mizoguchi K, Mitake S, Washimi Y, Tatsuoka Y, Fujimura H, Toda K, Kondo T, Nakashima K, Nomoto M, Uozumi T, Sato A, Matsuo H, Tsuruta K. Randomized placebo‐controlled trial of zonisamide in patients with Parkinson's disease. ACTA ACUST UNITED AC 2015. [DOI: 10.1111/ncn3.12026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Miho Murata
- Department of Neurology National Center Hospital National Center of Neurology and Psychiatry Tokyo Japan
| | - Kazuko Hasegawa
- Department of Neurology Sagamihara National Hospital SagamiharaTokyo Japan
| | - Ichiro Kanazawa
- International University of Health and Welfare Graduate School Tokyo Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Van der Schyf CJ. Rational drug discovery design approaches for treating Parkinson’s disease. Expert Opin Drug Discov 2015; 10:713-41. [DOI: 10.1517/17460441.2015.1041495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
43
|
Brimblecombe KR, Gracie CJ, Platt NJ, Cragg SJ. Gating of dopamine transmission by calcium and axonal N-, Q-, T- and L-type voltage-gated calcium channels differs between striatal domains. J Physiol 2015; 593:929-46. [PMID: 25533038 DOI: 10.1113/jphysiol.2014.285890] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/17/2014] [Indexed: 01/05/2023] Open
Abstract
KEY POINTS The voltage-gated Ca(2+) channels (VGCCs) that catalyse striatal dopamine transmission are critical to dopamine function and might prime subpopulations of neurons for parkinsonian degeneration. However, the VGCCs that operate on mesostriatal axons are incompletely defined; previous studies encompassed channels on striatal cholinergic interneurons that strongly influence dopamine transmission. We define that multiple types of axonal VGCCs operate that extend beyond classic presynaptic N/P/Q channels to include T- and L-types. We reveal differences in VGCC function between mouse axon types that in humans are vulnerable versus resistant to Parkinson's disease. We show for the first time that this is underpinned by different sensitivity of dopamine transmission to extracellular Ca(2+) and by different spatiotemporal intracellular Ca(2+) microdomains. These data define key principles of how Ca(2+) and VGCCs govern dopamine transmission in the healthy brain and reveal differences between neuron types that might contribute to vulnerability in disease. ABSTRACT The axonal voltage-gated Ca(2+) channels (VGCCs) that catalyse dopamine (DA) transmission are incompletely defined. Yet, they are critical to DA function and might prime subpopulations of DA neurons for parkinsonian degeneration. Previous studies of VGCCs will have encompassed those on striatal cholinergic interneurons, which strongly influence DA transmission. We identify which VGCCs on DA axons govern DA transmission, we determine their dynamic properties and reveal an underlying basis for differences between the caudate putamen (CPu) and nucleus accumbens (NAc). We detected DA release evoked electrically during nicotinic receptor blockade or optogenetically by light activation of channel rhodopsin-expressing DA axons in mouse striatal slices. Subtype-specific VGCC blockers indicated that N-, Q-, T- and L-VGCCs govern DA release in CPu, but in NAc, T and L-channels are relatively silent. The roles of the most dominant channels were inversely frequency-dependent, due to low-pass filtering of DA release by Ca(2+)-dependent relationships between initial release probability and short-term plasticity. Ca(2+) concentration-response curves revealed that differences between CPu and NAc were due to greater underlying Ca(2+) sensitivity of DA transmission from CPu axons. Functions for 'silent' L- and T-channels in NAc could be unmasked by elevating extracellular [Ca(2+)]. Furthermore, we identified a greater coupling between BAPTA-sensitive, fast Ca(2+) transients and DA transmission in CPu axons, and evidence for endogenous fast buffering of Ca(2+) in NAc. These data reveal that a range of VGCCs operate dynamically on DA axons, depending on local driving forces. Furthermore, they reveal dramatic differences in Ca(2+) handling between axonal subpopulations that show different vulnerability to parkinsonian degeneration.
Collapse
Affiliation(s)
- Katherine R Brimblecombe
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
44
|
Bosch-Bouju C, Smither RA, Hyland BI, Parr-Brownlie LC. Reduced reach-related modulation of motor thalamus neural activity in a rat model of Parkinson's disease. J Neurosci 2014; 34:15836-50. [PMID: 25429126 PMCID: PMC6608476 DOI: 10.1523/jneurosci.0893-14.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 10/01/2014] [Accepted: 10/04/2014] [Indexed: 02/06/2023] Open
Abstract
Motor thalamus (Mthal) is a key node in the corticobasal ganglia (BG) loop that controls complex, cognitive aspects of movement. In Parkinson's disease (PD), profound alterations in neuronal activity occur in BG nuclei and cortex. Because Mthal is located between these two structures, altered Mthal activity has been assumed to underlie the pathogenesis of PD motor deficits. However, to date, inconsistent changes in neuronal firing rate and pattern have been reported in parkinsonian animals. Moreover, although a distinct firing pattern of Mthal neurons, called low-threshold calcium spike bursts (LTS bursts), is observed in reduced preparations, it remains unknown whether they occur or what their role might be in behaving animals. We recorded Mthal spiking activity in control and unilateral 6-hydroxydopamine lesioned rats performing a skilled forelimb-reaching task. We show for the first time that Mthal firing rate in control rats is modulated in a temporally precise pattern during reach-to-grasp movements, with a peak at the time of the reach-end and troughs just before and after it. We identified LTS-like events on the basis of LTS burst characteristics. These were rare, but also modulated, decreasing in incidence just after reach-end. The inhibitory modulations in firing rate and LTS-like events were abolished in parkinsonian rats. These data confirm that nigrostriatal dopamine depletion is accompanied by profound and specific deficits in movement-related Mthal activity. These changes would severely impair Mthal contributions to motor program development in motor cortex and are likely to be an important factor underlying the movement deficits of PD.
Collapse
Affiliation(s)
| | - Roseanna A Smither
- Department of Physiology, Otago School of Medical Science, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| | - Brian I Hyland
- Department of Physiology, Otago School of Medical Science, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| | | |
Collapse
|
45
|
Yabe I, Ohta M, Egashira T, Sato K, Kano T, Hirotani M, Kunieda Y, Sasaki H. Effectiveness of zonisamide in a patient with Parkinson's disease and various levodopa-induced psychotic symptoms. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/ncn3.126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Ichiro Yabe
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Midori Ohta
- Rehabilitation Center; Wakkanai City Hospital; Wakkanai City Japan
| | - Toshiaki Egashira
- Department of Psychiatry; Wakkanai City Hospital; Wakkanai City Japan
| | - Kazunori Sato
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Takahiro Kano
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Makoto Hirotani
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Yasuyuki Kunieda
- Department of Internal Medicine; Wakkanai City Hospital; Wakkanai Japan
| | - Hidenao Sasaki
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| |
Collapse
|
46
|
Pan MK, Tai CH, Liu WC, Pei JC, Lai WS, Kuo CC. Deranged NMDAergic cortico-subthalamic transmission underlies parkinsonian motor deficits. J Clin Invest 2014; 124:4629-41. [PMID: 25202982 DOI: 10.1172/jci75587] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/31/2014] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is the most prevalent hypokinetic movement disorder, and symptomatic PD pathogenesis has been ascribed to imbalances between the direct and indirect pathways in the basal ganglia circuitry. Here, we applied glutamate receptor blockers to the subthalamic nucleus (STN) of parkinsonian rats and evaluated locomotor behaviors via single-unit and local-field recordings. Using this model, we found that inhibition of NMDAergic cortico-subthalamic transmission ameliorates parkinsonian motor deficits without eliciting any vivid turning behavior and abolishes electrophysiological abnormalities, including excessive subthalamic bursts, cortico-subthalamic synchronization, and in situ beta synchronization in both the motor cortex and STN. Premotor cortex stimulation revealed that cortico-subthalamic transmission is deranged in PD and directly responsible for the excessive stimulation-dependent bursts and time-locked spikes in the STN, explaining the genesis of PD-associated pathological bursts and synchronization, respectively. Moreover, application of a dopaminergic agent via a microinfusion cannula localized the therapeutic effect to the STN, without correcting striatal dopamine deficiency. Finally, optogenetic overactivation and synchronization of cortico-subthalamic transmission alone sufficiently and instantaneously induced parkinsonian-associated locomotor dysfunction in normal mice. In addition to the classic theory emphasizing the direct-indirect pathways, our data suggest that deranged cortico-subthalamic transmission via the NMDA receptor also plays a central role in the pathophysiology of parkinsonian motor deficits.
Collapse
|
47
|
Wilson D, Moehlis J. Locally optimal extracellular stimulation for chaotic desynchronization of neural populations. J Comput Neurosci 2014; 37:243-57. [PMID: 24899243 PMCID: PMC4159599 DOI: 10.1007/s10827-014-0499-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 03/02/2014] [Accepted: 03/10/2014] [Indexed: 10/26/2022]
Abstract
We use optimal control theory to design a methodology to find locally optimal stimuli for desynchronization of a model of neurons with extracellular stimulation. This methodology yields stimuli which lead to positive Lyapunov exponents, and hence desynchronizes a neural population. We analyze this methodology in the presence of interneuron coupling to make predictions about the strength of stimulation required to overcome synchronizing effects of coupling. This methodology suggests a powerful alternative to pulsatile stimuli for deep brain stimulation as it uses less energy than pulsatile stimuli, and could eliminate the time consuming tuning process.
Collapse
Affiliation(s)
- Dan Wilson
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA,
| | | |
Collapse
|
48
|
Gangarossa G, Laffray S, Bourinet E, Valjent E. T-type calcium channel Cav3.2 deficient mice show elevated anxiety, impaired memory and reduced sensitivity to psychostimulants. Front Behav Neurosci 2014; 8:92. [PMID: 24672455 PMCID: PMC3957728 DOI: 10.3389/fnbeh.2014.00092] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/03/2014] [Indexed: 01/28/2023] Open
Abstract
The fine-tuning of neuronal excitability relies on a tight control of Ca2+ homeostasis. The low voltage-activated (LVA) T-type calcium channels (Cav3.1, Cav3.2 and Cav3.3 isoforms) play a critical role in regulating these processes. Despite their wide expression throughout the central nervous system, the implication of T-type Cav3.2 isoform in brain functions is still poorly characterized. Here, we investigate the effect of genetic ablation of this isoform in affective disorders, including anxiety, cognitive functions as well as sensitivity to drugs of abuse. Using a wide range of behavioral assays we show that genetic ablation of the cacna1h gene results in an anxiety-like phenotype, whereas novelty-induced locomotor activity is unaffected. Deletion of the T-type channel Cav3.2 also triggers impairment of hippocampus-dependent recognition memories. Acute and sensitized hyperlocomotion induced by d-amphetamine and cocaine are dramatically reduced in T-type Cav3.2 deficient mice. In addition, the administration of the T-type blocker TTA-A2 prevented the expression of locomotor sensitization observed in wildtype mice. In conclusion, our data reveal that physiological activity of this specific Ca2+ channel is required for affective and cognitive behaviors. Moreover, our work highlights the interest of T-type channel blockers as therapeutic strategies to reverse drug-associated alterations.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- Institut de Génomique Fonctionnelle, CNRS UMR-5203, Montpellier, France ; INSERM U661, Montpellier, France ; Universités de Montpellier 1 and 2 UMR-5203, Montpellier, France
| | - Sophie Laffray
- Institut de Génomique Fonctionnelle, CNRS UMR-5203, Montpellier, France ; INSERM U661, Montpellier, France ; Universités de Montpellier 1 and 2 UMR-5203, Montpellier, France ; Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle Montpellier, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle, CNRS UMR-5203, Montpellier, France ; INSERM U661, Montpellier, France ; Universités de Montpellier 1 and 2 UMR-5203, Montpellier, France ; Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle Montpellier, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle, CNRS UMR-5203, Montpellier, France ; INSERM U661, Montpellier, France ; Universités de Montpellier 1 and 2 UMR-5203, Montpellier, France
| |
Collapse
|
49
|
Yang YC, Tai CH, Pan MK, Kuo CC. The T-type calcium channel as a new therapeutic target for Parkinson's disease. Pflugers Arch 2014; 466:747-55. [PMID: 24531801 DOI: 10.1007/s00424-014-1466-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is one of the most prevalent movement disorder caused by degeneration of the dopaminergic neurons in substantia nigra pars compacta. Deep brain stimulation (DBS) at the subthalamic nucleus (STN) has been a new and effective treatment of PD. It is interesting how a neurological disorder caused by the deficiency of a specific chemical substance (i.e., dopamine) from one site could be so successfully treated by a pure physical maneuver (i.e., DBS) at another site. STN neurons could discharge in the single-spike or the burst modes. A significant increase in STN burst discharges has been unequivocally observed in dopamine-deprived conditions such as PD, and was recently shown to have a direct causal relation with parkinsonian symptoms. The occurrence of burst discharges in STN requires enough available T-type Ca(2+) currents, which could bring the relatively negative membrane potential to the threshold of firing Na(+) spikes. DBS, by injection of negative currents into the extracellular space, most likely would depolarize the STN neuron and then inactivate the T-type Ca(2+) channel. Burst discharges are thus decreased and parkinsonian locomotor deficits ameliorated. Conversely, injection of positive currents into STN itself could induce parkinsonian locomotor deficits in animals without dopaminergic lesions. Local application of T-type Ca(2+) channel blockers into STN would also dramatically decrease the burst discharges and improve parkinsonian locomotor symptoms. Notably, zonisamide, which could inhibit T-type Ca(2+) currents in STN, has been shown to benefit PD patients in a clinical trial. From the pathophysiological perspectives, PD can be viewed as a prototypical disorder of "brain arrhythmias". Modulation of relevant ion channels by physical or chemical maneuvers may be important therapeutic considerations for PD and other diseases related to deranged neural rhythms.
Collapse
Affiliation(s)
- Ya-Chin Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | |
Collapse
|
50
|
Cataldi M. The changing landscape of voltage-gated calcium channels in neurovascular disorders and in neurodegenerative diseases. Curr Neuropharmacol 2013; 11:276-97. [PMID: 24179464 PMCID: PMC3648780 DOI: 10.2174/1570159x11311030004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/02/2013] [Accepted: 02/14/2013] [Indexed: 12/12/2022] Open
Abstract
It is a common belief that voltage-gated calcium channels (VGCC) cannot carry toxic amounts of Ca2+ in neurons. Also, some of them as L-type channels are essential for Ca2+-dependent regulation of prosurvival gene-programs. However, a wealth of data show a beneficial effect of drugs acting on VGCCs in several neurodegenerative and neurovascular diseases. In the present review, we explore several mechanisms by which the “harmless” VGCCs may become “toxic” for neurons. These mechanisms could explain how, though usually required for neuronal survival, VGCCs may take part in neurodegeneration. We will present evidence showing that VGCCs can carry toxic Ca2+ when: a) their density or activity increases because of aging, chronic hypoxia or exposure to β-amyloid peptides or b) Ca2+-dependent action potentials carry high Ca2+ loads in pacemaker neurons. Besides, we will examine conditions in which VGCCs promote neuronal cell death without carrying excess Ca2+. This can happen, for instance, when they carry metal ions into the neuronal cytoplasm or when a pathological decrease in their activity weakens Ca2+-dependent prosurvival gene programs. Finally, we will explore the role of VGCCs in the control of nonneuronal cells that take part to neurodegeneration like those of the neurovascular unit or of microglia.
Collapse
Affiliation(s)
- Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Italy
| |
Collapse
|