1
|
Enger R, Heuser K. Astrocytes as critical players of the fine balance between inhibition and excitation in the brain: spreading depolarization as a mechanism to curb epileptic activity. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1360297. [PMID: 38405021 PMCID: PMC10884165 DOI: 10.3389/fnetp.2024.1360297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 02/27/2024]
Abstract
Spreading depolarizations (SD) are slow waves of complete depolarization of brain tissue followed by neuronal silencing that may play a role in seizure termination. Even though SD was first discovered in the context of epilepsy research, the link between SD and epileptic activity remains understudied. Both seizures and SD share fundamental pathophysiological features, and recent evidence highlights the frequent occurrence of SD in experimental seizure models. Human data on co-occurring seizures and SD are limited but suggestive. This mini-review addresses possible roles of SD during epileptiform activity, shedding light on SD as a potential mechanism for terminating epileptiform activity. A common denominator for many forms of epilepsy is reactive astrogliosis, a process characterized by morphological and functional changes to astrocytes. Data suggest that SD mechanisms are potentially perturbed in reactive astrogliosis and we propose that this may affect seizure pathophysiology.
Collapse
Affiliation(s)
- Rune Enger
- Letten Centre and GliaLab, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kjell Heuser
- Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
2
|
Gray MM, Naik A, Ebner TJ, Carter RE. Altered brain state during episodic dystonia in tottering mice decouples primary motor cortex from limb kinematics. DYSTONIA 2023; 2:10974. [PMID: 37800168 PMCID: PMC10554815 DOI: 10.3389/dyst.2023.10974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Episodic Ataxia Type 2 (EA2) is a rare neurological disorder caused by a mutation in the CACNA1A gene, encoding the P/Q-type voltage-gated Ca2+ channel important for neurotransmitter release. Patients with this channelopathy exhibit both cerebellar and cerebral pathologies, suggesting the condition affects both regions. The tottering (tg/tg) mouse is the most commonly used EA2 model due to an orthologous mutation in the cacna1a gene. The tg/tg mouse has three prominent behavioral phenotypes: a dramatic episodic dystonia; absence seizures with generalized spike and wave discharges (GSWDs); and mild ataxia. We previously observed a novel brain state, transient low-frequency oscillations (LFOs) in the cerebellum and cerebral cortex under anesthesia. In this study, we examine the relationships among the dystonic attack, GSWDs, and LFOs in the cerebral cortex. Previous studies characterized LFOs in the motor cortex of anesthetized tg/tg mice using flavoprotein autofluorescence imaging testing the hypothesis that LFOs provide a mechanism for the paroxysmal dystonia. We sought to obtain a more direct understanding of motor cortex (M1) activity during the dystonic episodes. Using two-photon Ca2+ imaging to investigate neuronal activity in M1 before, during, and after the dystonic attack, we show that there is not a significant change in the activity of M1 neurons from baseline through the attack. We also conducted simultaneous, multi-electrode recordings to further understand how M1 cellular activity and local field potentials change throughout the progression of the dystonic attack. Neither putative pyramidal nor inhibitory interneuron firing rate changed during the dystonic attack. However, we did observe a near complete loss of GSWDs during the dystonic attack in M1. Finally, using spike triggered averaging to align simultaneously recorded limb kinematics to the peak Ca2+ response, and vice versa, revealed a reduction in the spike triggered average during the dystonic episodes. Both the loss of GSWDs and the reduction in the coupling suggest that, during the dystonic attack, M1 is effectively decoupled from other structures. Overall, these results indicate that the attack is not initiated or controlled in M1, but elsewhere in the motor circuitry. The findings also highlight that LFOs, GSWDs, and dystonic attacks represent three brain states in tg/tg mice.
Collapse
Affiliation(s)
- Madelyn M Gray
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Anant Naik
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Russell E Carter
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
3
|
Ren L, Gang X, Yang S, Sun M, Wang G. A new perspective of hypothalamic disease: Shapiro's syndrome. Front Neurol 2022; 13:911332. [PMID: 35968294 PMCID: PMC9372501 DOI: 10.3389/fneur.2022.911332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Shapiro's syndrome (SS) is characterized by spontaneous periodic hypothermia. It occurs to patients regardless of age or sex. To date, <60 cases have been reported worldwide. Current knowledge of the disease is limited to clinical feature since the pathogenesis and etiology are still controversial. In this review, the clinical characteristics, pathological mechanism, and possible etiology of the syndrome were reviewed to improve the clinical understanding of the disease.
Collapse
Affiliation(s)
- Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Shuo Yang
- Department of Clinical Nutrition, The First Hospital of Jilin University, Changchun, China
| | - Meixin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Guixia Wang
| |
Collapse
|
4
|
Snell HD, Vitenzon A, Tara E, Chen C, Tindi J, Jordan BA, Khodakhah K. Mechanism of stress-induced attacks in an episodic neurologic disorder. SCIENCE ADVANCES 2022; 8:eabh2675. [PMID: 35442745 PMCID: PMC9020779 DOI: 10.1126/sciadv.abh2675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/02/2022] [Indexed: 05/21/2023]
Abstract
Stress is the most common trigger among episodic neurologic disorders. In episodic ataxia type 2 (EA2), physical or emotional stress causes episodes of severe motor dysfunction that manifest as ataxia and dystonia. We used the tottering (tg/tg) mouse, a faithful animal model of EA2, to dissect the mechanisms underlying stress-induced motor attacks. We find that in response to acute stress, activation of α1-adrenergic receptors (α1-Rs) on Purkinje cells by norepinephrine leads to their erratic firing and consequently motor attacks. We show that norepinephrine induces erratic firing of Purkinje cells by disrupting their spontaneous intrinsic pacemaking via a casein kinase 2 (CK2)-dependent signaling pathway, which likely reduces the activity of calcium-dependent potassium channels. Moreover, we report that disruption of this signaling cascade at a number of nodes prevents stress-induced attacks in the tottering mouse. Together, our results suggest that norepinephrine and CK2 are required for the initiation of stress-induced attacks in EA2 and provide previously unidentified targets for therapeutic intervention.
Collapse
Affiliation(s)
- Heather D. Snell
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ariel Vitenzon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Esra Tara
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chris Chen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jaafar Tindi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bryen A. Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Psychiatry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Psychiatry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
5
|
Liu R, Sun L, Wang Y, Jia M, Wang Q, Cai X, Wu J. Double-edged Role of K Na Channels in Brain Tuning: Identifying Epileptogenic Network Micro-Macro Disconnection. Curr Neuropharmacol 2022; 20:916-928. [PMID: 34911427 PMCID: PMC9881102 DOI: 10.2174/1570159x19666211215104829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
Epilepsy is commonly recognized as a disease driven by generalized hyperexcited and hypersynchronous neural activity. Sodium-activated potassium channels (KNa channels), which are encoded by the Slo 2.2 and Slo 2.1 genes, are widely expressed in the central nervous system and considered as "brakes" to adjust neuronal adaptation through regulating action potential threshold or after-hyperpolarization under physiological condition. However, the variants in KNa channels, especially gain-of-function variants, have been found in several childhood epileptic conditions. Most previous studies focused on mapping the epileptic network on the macroscopic scale while ignoring the value of microscopic changes. Notably, paradoxical role of KNa channels working on individual neuron/microcircuit and the macroscopic epileptic expression highlights the importance of understanding epileptogenic network through combining microscopic and macroscopic methods. Here, we first illustrated the molecular and physiological function of KNa channels on preclinical seizure models and patients with epilepsy. Next, we summarized current hypothesis on the potential role of KNa channels during seizures to provide essential insight into what emerged as a micro-macro disconnection at different levels. Additionally, we highlighted the potential utility of KNa channels as therapeutic targets for developing innovative anti-seizure medications.
Collapse
Affiliation(s)
- Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Lei Sun
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Meng Jia
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiang Cai
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,Address correspondence to these authors at the Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Tel: +0086-18062552085; E-mail: Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China; Tel: +0086-13319285082; E-mail:
| | - Jianping Wu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China;,Address correspondence to these authors at the Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Tel: +0086-18062552085; E-mail: Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China; Tel: +0086-13319285082; E-mail:
| |
Collapse
|
6
|
Tian WT, Zhan FX, Liu ZH, Liu Z, Liu Q, Guo XN, Zhou ZW, Wang SG, Liu XR, Jiang H, Li XH, Zhao GH, Li HY, Tang JG, Bi GH, Zhong P, Yin XM, Liu TT, Ni RL, Zheng HR, Liu XL, Qian XH, Wu JY, Cao YW, Zhang C, Liu SH, Wu YY, Wang QF, Xu T, Hou WZ, Li ZY, Ke HY, Zhu ZY, Zheng L, Wang T, Rong TY, Wu L, Zhang Y, Fang K, Wang ZH, Zhang YK, Zhang M, Zhao YW, Tang BS, Luan XH, Huang XJ, Cao L. TMEM151A Variants Cause Paroxysmal Kinesigenic Dyskinesia: A Large-Sample Study. Mov Disord 2022; 37:545-552. [PMID: 34820915 DOI: 10.1002/mds.28865] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Paroxysmal kinesigenic dyskinesia (PKD) is the most common type of paroxysmal dyskinesias. Only one-third of PKD patients are attributed to proline-rich transmembrane protein 2 (PRRT2) mutations. OBJECTIVE We aimed to explore the potential causative gene for PKD. METHODS A cohort of 196 PRRT2-negative PKD probands were enrolled for whole-exome sequencing (WES). Gene Ranking, Identification and Prediction Tool, a method of case-control analysis, was applied to identify the candidate genes. Another 325 PRRT2-negative PKD probands were subsequently screened with Sanger sequencing. RESULTS Transmembrane Protein 151 (TMEM151A) variants were mainly clustered in PKD patients compared with the control groups. 24 heterozygous variants were detected in 25 of 521 probands (frequency = 4.80%), including 18 missense and 6 nonsense mutations. In 29 patients with TMEM151A variants, the ratio of male to female was 2.63:1 and the mean age of onset was 12.93 ± 3.15 years. Compared with PRRT2 mutation carriers, TMEM151A-related PKD were more common in sporadic PKD patients with pure phenotype. There was no significant difference in types of attack and treatment outcome between TMEM151A-positive and PRRT2-positive groups. CONCLUSIONS We consolidated mutations in TMEM151A causing PKD with the aid of case-control analysis of a large-scale WES data, which broadens the genotypic spectrum of PKD. TMEM151A-related PKD were more common in sporadic cases and tended to present as pure phenotype with a late onset. Extensive functional studies are needed to enhance our understanding of the pathogenesis of TMEM151A-related PKD. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Wo-Tu Tian
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Fei-Xia Zhan
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen-Hua Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhe Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Laboratory of Clinical Genetics, Medical Science Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qing Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xia-Nan Guo
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, Dalian, China
| | - Zai-Wei Zhou
- Shanghai Xunyin Biotechnology Co., Ltd., Shanghai, China
| | - Shi-Ge Wang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Rong Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Institute of Neuroscience of The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong Jiang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xun-Hua Li
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guo-Hua Zhao
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Hai-Yan Li
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Anyang People's Hospital, Anyang, China
| | - Jian-Guang Tang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guang-Hui Bi
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Dongying People's Hospital, Dongying, China
| | - Ping Zhong
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Xiao-Meng Yin
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tao-Tao Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science & Technology, Huainan, China
| | - Rui-Long Ni
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science & Technology, Huainan, China
| | - Hao-Ran Zheng
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science & Technology, Huainan, China
| | - Xiao-Li Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Xiao-Hang Qian
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Ying Wu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Yu-Wen Cao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Chao Zhang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Shi-Hua Liu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Ying-Ying Wu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Qun-Feng Wang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Ting Xu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Wen-Zhe Hou
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Zi-Yi Li
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-Yi Ke
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yu Zhu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Lan Zheng
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Tian Wang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Yi Rong
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai, China
| | - Li Wu
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kan Fang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhan-Hang Wang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Ya-Kun Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mei Zhang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science & Technology, Huainan, China
| | - Yu-Wu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Bei-Sha Tang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xing-Hua Luan
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
| | - Xiao-Jun Huang
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Cao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- China Paroxysmal Dyskinesia Collaborative Group (CPDCG), Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science & Technology, Huainan, China
| |
Collapse
|
7
|
Inherited Developmental and Epileptic Encephalopathies. Neurol Int 2021; 13:555-568. [PMID: 34842787 PMCID: PMC8628919 DOI: 10.3390/neurolint13040055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 01/13/2023] Open
Abstract
Epileptic encephalopathies often have a genetic etiology. The epileptic activity itself exerts a direct detrimental effect on neurodevelopment, which may add to the cognitive impairment induced by the underlying mutation (“developmental and epileptic encephalopathy”). The focus of this review is on inherited syndromes. The phenotypes of genetic disorders affecting ion channels, metabolic signalling, membrane trafficking and exocytosis, cell adhesion, cell growth and proliferation are discussed. Red flags suggesting family of genes or even specific genes are highlighted. The knowledge of the phenotypical spectrum can indeed prompt the clinician to suspect specific etiologies, expediting the diagnosis.
Collapse
|
8
|
Roy PK, Rajesh Y, Mandal M. Therapeutic targeting of membrane-associated proteins in central nervous system tumors. Exp Cell Res 2021; 406:112760. [PMID: 34339674 DOI: 10.1016/j.yexcr.2021.112760] [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: 04/30/2021] [Revised: 06/28/2021] [Accepted: 07/28/2021] [Indexed: 12/09/2022]
Abstract
The activity of the most complex system, the central nervous system (CNS) is profoundly regulated by a huge number of membrane-associated proteins (MAP). A minor change stimulates immense chemical changes and the elicited response is organized by MAP, which acts as a receptor of that chemical or channel enabling the flow of ions. Slight changes in the activity or expression of these MAPs lead to severe consequences such as cognitive disorders, memory loss, or cancer. CNS tumors are heterogeneous in nature and hard-to-treat due to random mutations in MAPs; like as overexpression of EGFRvIII/TGFβR/VEGFR, change in adhesion molecules α5β3 integrin/SEMA3A, imbalance in ion channel proteins, etc. Extensive research is under process for developing new therapeutic approaches using these proteins such as targeted cytotoxic radiotherapy, drug-delivery, and prodrug activation, blocking of receptors like GluA1, developing viral vector against cell surface receptor. The combinatorial approach of these strategies along with the conventional one might be more potential. Henceforth, our review focuses on in-depth analysis regarding MAPs aiming for a better understanding for developing an efficient therapeutic approach for targeting CNS tumors.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India
| | - Yetirajam Rajesh
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India.
| |
Collapse
|
9
|
Li W, Zhao W, Wang J, Zhang X, Qian X, Gu R, He G. Identification of a novel variant p.Ser606Gly in SCN3A associated with childhood absence epilepsy. Epilepsy Res 2021; 175:106682. [PMID: 34102392 DOI: 10.1016/j.eplepsyres.2021.106682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Sodium (Na+) channels are the basis for action potential generation and propagation, which play a key role in the regulation of neuronal excitability. SCN3A is a gene encoding for sodium channel protein type 3 subunit alpha (or known as Nav1.3). This study aimed to explore SCN3A genetic variants in a cohort of childhood absence epilepsy (CAE) via whole exome sequencing. A novel SCN3A missense variant (c.A1816G, p.Ser606Gly) was identified in a patient with CAE. This variant had not been reported in both 1000G and ExAC databases. Bioinformatics analysis revealed that this variant was pathogenic and could transform the protein structure of Nav1.3. The reported phenotypes of SCN3A-related central nerve system disorders included multiple seizure types, polymicrogyria and different degrees of developmental delay/intellectual disability. The patient with p.Ser606Gly variant exhibited typical absence seizures. The MRI and CT scan results were normal, and EEG showed that 3-Hz spike-slow wave discharges. In conclusion, our findings not only broaden the pathogenic spectrum of SCN3A, but also extend the clinical phenotypes of SCN3A-related CAE.
Collapse
Affiliation(s)
- Wei Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Wenli Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jing Wang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xiaoli Zhang
- Department of Neurology, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xinlai Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Renjun Gu
- Department of Neurology, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| | - Guoyang He
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
10
|
Danti FR, Invernizzi F, Moroni I, Garavaglia B, Nardocci N, Zorzi G. Pediatric Paroxysmal Exercise-Induced Neurological Symptoms: Clinical Spectrum and Diagnostic Algorithm. Front Neurol 2021; 12:658178. [PMID: 34140924 PMCID: PMC8203909 DOI: 10.3389/fneur.2021.658178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Paroxysmal exercise-induced neurological symptoms (PENS) encompass a wide spectrum of clinical phenomena commonly presenting during childhood and characteristically elicited by physical exercise. Interestingly, few shared pathogenetic mechanisms have been identified beyond the well-known entity of paroxysmal exercise-induced dyskinesia, PENS could be part of more complex phenotypes including neuromuscular, neurodegenerative, and neurometabolic disease, epilepsies, and psychogenetic disorders. The wide and partially overlapping phenotypes and the genetic heterogeneity make the differential diagnosis frequently difficult and delayed; however, since some of these disorders may be treatable, a prompt diagnosis is mandatory. Therefore, an accurate characterization of these symptoms is pivotal for orienting more targeted biochemical, radiological, neurophysiological, and genetic investigations and finally treatment. In this article, we review the clinical, genetic, pathophysiologic, and therapeutic landscape of paroxysmal exercise induced neurological symptoms, focusing on phenomenology and differential diagnosis.
Collapse
Affiliation(s)
- Federica Rachele Danti
- Unit of Child Neurology, Department of Pediatric Neuroscience, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Invernizzi
- Unit of Medical Genetics and Neurogenetics, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan, Italy
| | - Isabella Moroni
- Unit of Child Neurology, Department of Pediatric Neuroscience, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Barbara Garavaglia
- Unit of Medical Genetics and Neurogenetics, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico C. Besta, Milan, Italy
| | - Nardo Nardocci
- Unit of Child Neurology, Department of Pediatric Neuroscience, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanna Zorzi
- Unit of Child Neurology, Department of Pediatric Neuroscience, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
11
|
|
12
|
Adult occipital lobe epilepsy: 12-years on. J Neurol 2021; 268:3926-3934. [PMID: 33900448 DOI: 10.1007/s00415-021-10557-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Occipital lobe epilepsies (OLE) comprise 5-10% of focal epilepsies in surgical and paediatric series; with little data from adult medical cohorts. This longitudinal study examined OLE patients, to characterise prevalence, semiology, co-morbidity and prognosis in a neurology outpatient setting. METHODS 24 adult OLE patients identified over 12 months from 1548 patients in a neurologist's service were followed over 12 years. RESULTS 92% of these OLE patients had simple visual hallucinations, misdiagnosed in 40% of cases. 75% had co-morbid interictal migraine and 38% had visual field defects. Only 33% achieved long-term remission, and only 2 /10 (20%) of OLE patients with a structural aetiology were seizure-free. The two patients with migralepsy achieved remission. CONCLUSION Adult OLE accounted for 7.7% of focal epilepsies in this cohort, misdiagnosed or misclassified in 40%. Most patients had co-existing migraine. A minority had migralepsy characterised by a longer aura and good prognosis. Remission rates were lower than that of childhood OLE and general adult epilepsy populations, strengthening the argument for considering epilepsy surgery in drug-resistant OLE patients with a structural cause. Precision medicine will potentially refine diagnosis and management in those OLE patients without an identified cause but is predicated on accurate clinical phenotyping.
Collapse
|
13
|
Evidence Supporting the Regulatory Relationships through a Paracrine Pathway between the Sternum and Pectoral Muscles in Ducks. Genes (Basel) 2021; 12:genes12040463. [PMID: 33804959 PMCID: PMC8063953 DOI: 10.3390/genes12040463] [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: 01/28/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Muscles and bones are anatomically closely linked, and they can conduct communication by mechanical and chemical signals. However, the specific regulatory mechanism between the pectoral muscle and sternum in birds was largely unknown. The present study explored the potential relationship between them in ducks. The result of the sections showed that more nuclei in proliferate states were observed in the pectoral muscle fibers attached to the calcified sternum, than those attached to the un-calcified sternum. The RNA-seq identified 328 differentially expressed genes (DEGs) in the sternum between the calcified and un-calcified groups. Gene ontology (GO) showed that the DEGs were mainly enriched in pathways associated with calcification. In addition, DEGs in the muscles between the calcified and un-calcified sternum groups were mainly annotated to signal transduction receptor pathways. The expression patterns of genes encoding for secreted proteins, in bone (CXCL12, BMP7 and CTSK) and muscle (LGI1), were clustered with muscle development (MB) and bone calcification (KCNA1, OSTN, COL9A3, and DCN) related genes, respectively, indicating the regulatory relationships through a paracrine pathway existing between the sternum and pectoral muscles in ducks. Together, we demonstrated that the pectoral muscle development was affected by the sternal ossification states in ducks. The VEGFA, CXCL12, SPP1, NOG, and BMP7 were possibly the key genes to participate in the ossification of the duck sternum. We firstly listed evidence supporting the regulatory relationships through a paracrine pathway between the sternum and pectoral muscles in ducks, which provided scientific data for the study of the synergistic development of bone and skeletal muscle.
Collapse
|
14
|
Niklas B, Jankowska M, Gordon D, Béress L, Stankiewicz M, Nowak W. Interactions of Sea Anemone Toxins with Insect Sodium Channel-Insights from Electrophysiology and Molecular Docking Studies. Molecules 2021; 26:molecules26051302. [PMID: 33670972 PMCID: PMC7957711 DOI: 10.3390/molecules26051302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023] Open
Abstract
Animal venoms are considered as a promising source of new drugs. Sea anemones release polypeptides that affect electrical activity of neurons of their prey. Voltage dependent sodium (Nav) channels are the common targets of Av1, Av2, and Av3 toxins from Anemonia viridis and CgNa from Condylactis gigantea. The toxins bind to the extracellular side of a channel and slow its fast inactivation, but molecular details of the binding modes are not known. Electrophysiological measurements on Periplaneta americana neuronal preparation revealed differences in potency of these toxins to increase nerve activity. Av1 and CgNa exhibit the strongest effects, while Av2 the weakest effect. Extensive molecular docking using a modern SMINA computer method revealed only partial overlap among the sets of toxins’ and channel’s amino acid residues responsible for the selectivity and binding modes. Docking positions support earlier supposition that the higher neuronal activity observed in electrophysiology should be attributed to hampering the fast inactivation gate by interactions of an anemone toxin with the voltage driven S4 helix from domain IV of cockroach Nav channel (NavPaS). Our modelling provides new data linking activity of toxins with their mode of binding in site 3 of NavPaS channel.
Collapse
Affiliation(s)
- Beata Niklas
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100 Torun, Poland
- Correspondence: (B.N.); (W.N.)
| | - Milena Jankowska
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (M.J.); (M.S.)
| | - Dalia Gordon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - László Béress
- Department of Internal Medicine, Clinic of Immunology, Division of Experimental and Clinical Peptide Research, Hannover Medical School, 30625 Hannover, Germany;
| | - Maria Stankiewicz
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (M.J.); (M.S.)
| | - Wieslaw Nowak
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100 Torun, Poland
- Correspondence: (B.N.); (W.N.)
| |
Collapse
|
15
|
Smith RS, Walsh CA. Ion Channel Functions in Early Brain Development. Trends Neurosci 2020; 43:103-114. [PMID: 31959360 PMCID: PMC7092371 DOI: 10.1016/j.tins.2019.12.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
During prenatal brain development, ion channels are ubiquitous across several cell types, including progenitor cells and migrating neurons but their function has not been clear. In the past, ion channel dysfunction has been primarily studied in the context of postnatal, differentiated neurons that fire action potentials - notably ion channels mutated in the epilepsies - yet data now support a surprising role in prenatal human brain disorders as well. Modern gene discovery approaches have identified defective ion channels in individuals with cerebral cortex malformations, which reflect abnormalities in early-to-middle stages of embryonic development (prior to ubiquitous action potentials). These human genetics studies and recent in utero animal modeling work suggest that precise control of ionic flux (calcium, sodium, and potassium) contributes to in utero developmental processes such as neural proliferation, migration, and differentiation.
Collapse
Affiliation(s)
- Richard S Smith
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Bartolini E, Campostrini R, Kiferle L, Pradella S, Rosati E, Chinthapalli K, Palumbo P. Epilepsy and brain channelopathies from infancy to adulthood. Neurol Sci 2019; 41:749-761. [PMID: 31838630 DOI: 10.1007/s10072-019-04190-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/06/2019] [Indexed: 01/04/2023]
Abstract
Genetic brain channelopathies result from inherited or de novo mutations of genes encoding ion channel subunits within the central nervous system. Most neurological channelopathies arise in childhood with paroxysmal or episodic symptoms, likely because of a transient impairment of homeostatic mechanisms regulating membrane excitability, and the prototypical expression of this impairment is epilepsy. Migraine, episodic ataxia and alternating hemiplegia can also occur, as well as chronic phenotypes, such as spinocerebellar ataxias, intellectual disability and autism spectrum disorder. Voltage-gated and ligand-gated channels may be involved. In most cases, a single gene may be associated with a phenotypical spectrum that shows variable expressivity. Different clinical features may arise at different ages and the adult phenotype may be remarkably modified from the syndrome onset in childhood or adolescence. Recognizing the prominent phenotypical traits of brain channelopathies is essential to perform appropriate diagnostic investigations and to provide the better care not only in the paediatric setting but also for adult patients and their caregivers. Herein, we provide an overview of genetic brain channelopathies associated with epilepsy, highlight the different molecular mechanisms and describe the different clinical characteristics which may prompt the clinician to suspect specific syndromes and to possibly establish tailored treatments.
Collapse
Affiliation(s)
- Emanuele Bartolini
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy.
| | - Roberto Campostrini
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Lorenzo Kiferle
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Silvia Pradella
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Eleonora Rosati
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | | | - Pasquale Palumbo
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| |
Collapse
|
17
|
Liu C, Li T, Chen J. Role of High‐Throughput Electrophysiology in Drug Discovery. ACTA ACUST UNITED AC 2019; 87:e69. [DOI: 10.1002/cpph.69] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Chang Liu
- Department of Biochemical and Cellular PharmacologyGenentech Early Research and Development, Genentech South San Francisco California
| | - Tianbo Li
- Department of Biochemical and Cellular PharmacologyGenentech Early Research and Development, Genentech South San Francisco California
| | - Jun Chen
- Department of Biochemical and Cellular PharmacologyGenentech Early Research and Development, Genentech South San Francisco California
| |
Collapse
|
18
|
Brusich DJ, Spring AM, James TD, Yeates CJ, Helms TH, Frank CA. Drosophila CaV2 channels harboring human migraine mutations cause synapse hyperexcitability that can be suppressed by inhibition of a Ca2+ store release pathway. PLoS Genet 2018; 14:e1007577. [PMID: 30080864 PMCID: PMC6095605 DOI: 10.1371/journal.pgen.1007577] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/16/2018] [Accepted: 07/20/2018] [Indexed: 11/28/2022] Open
Abstract
Gain-of-function mutations in the human CaV2.1 gene CACNA1A cause familial hemiplegic migraine type 1 (FHM1). To characterize cellular problems potentially triggered by CaV2.1 gains of function, we engineered mutations encoding FHM1 amino-acid substitutions S218L (SL) and R192Q (RQ) into transgenes of Drosophila melanogaster CaV2/cacophony. We expressed the transgenes pan-neuronally. Phenotypes were mild for RQ-expressing animals. By contrast, single mutant SL- and complex allele RQ,SL-expressing animals showed overt phenotypes, including sharply decreased viability. By electrophysiology, SL- and RQ,SL-expressing neuromuscular junctions (NMJs) exhibited enhanced evoked discharges, supernumerary discharges, and an increase in the amplitudes and frequencies of spontaneous events. Some spontaneous events were gigantic (10-40 mV), multi-quantal events. Gigantic spontaneous events were eliminated by application of TTX-or by lowered or chelated Ca2+-suggesting that gigantic events were elicited by spontaneous nerve firing. A follow-up genetic approach revealed that some neuronal hyperexcitability phenotypes were reversed after knockdown or mutation of Drosophila homologs of phospholipase Cβ (PLCβ), IP3 receptor, or ryanodine receptor (RyR)-all factors known to mediate Ca2+ release from intracellular stores. Pharmacological inhibitors of intracellular Ca2+ store release produced similar effects. Interestingly, however, the decreased viability phenotype was not reversed by genetic impairment of intracellular Ca2+ release factors. On a cellular level, our data suggest inhibition of signaling that triggers intracellular Ca2+ release could counteract hyperexcitability induced by gains of CaV2.1 function.
Collapse
Affiliation(s)
- Douglas J. Brusich
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
| | - Ashlyn M. Spring
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States of America
| | - Thomas D. James
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States of America
| | - Catherine J. Yeates
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States of America
| | - Timothy H. Helms
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
19
|
Tu ML, Fang YW, Leu JG, Tsai MH. An atypical presentation of high potassium renal secretion rate in a patient with thyrotoxic periodic paralysis: a case report. BMC Nephrol 2018; 19:160. [PMID: 29973184 PMCID: PMC6031107 DOI: 10.1186/s12882-018-0971-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023] Open
Abstract
Background Hypokalemia is one of the most common clinical electrolyte imbalance problems, and thyrotoxic periodic paralysis (TPP) is a leading cause of presentation to the emergency department. Low renal potassium secretion rates, a normal acid–base balance in the blood, and hyperthyroidism are the hallmarks of suspected TPP. Case presentation Here we report the case of a 36-year-old man who presented to the emergency department with a sudden onset of acute muscle weakness at 5 h prior to admission. Biochemistry tests revealed hypokalemia with hyperthyroidism and renal potassium wasting. TPP was initially not favored due to the presence of renal potassium wasting. However, his serum potassium level rebounded rapidly within several hours after potassium supplementation, indicating that the intracellular shifting of potassium ions was the main etiology for his hypokalemia. The early stage of TPP development may have contributed to this paradox. Conclusion Therefore, it is premature to rule out TPP based on the presentation of high renal potassium secretion rates alone. This finding may result in an incorrect impression being made in the early stage of TTP and may consequently lead to an inappropriate potassium supplementation policy.
Collapse
Affiliation(s)
- Mei-Lan Tu
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, 95, Wen-Chang Rd, Shih-Lin, Taipei, 111, Taiwan (R.O.C.)
| | - Yu-Wei Fang
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, 95, Wen-Chang Rd, Shih-Lin, Taipei, 111, Taiwan (R.O.C.).,Fu-Jen Catholic University School of Medicine, Taipei, Taiwan (R.O.C.)
| | - Jyh-Gang Leu
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, 95, Wen-Chang Rd, Shih-Lin, Taipei, 111, Taiwan (R.O.C.).,Fu-Jen Catholic University School of Medicine, Taipei, Taiwan (R.O.C.)
| | - Ming-Hsien Tsai
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, 95, Wen-Chang Rd, Shih-Lin, Taipei, 111, Taiwan (R.O.C.). .,Fu-Jen Catholic University School of Medicine, Taipei, Taiwan (R.O.C.).
| |
Collapse
|
20
|
Guterman EL, Yurgionas B, Nelson AB. Pearls & Oy-sters: Episodic ataxia type 2: Case report and review of the literature. Neurology 2018; 86:e239-41. [PMID: 27272039 DOI: 10.1212/wnl.0000000000002743] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Elan L Guterman
- From the Department of Neurology, University of California, San Francisco.
| | - Brian Yurgionas
- From the Department of Neurology, University of California, San Francisco
| | - Alexandra B Nelson
- From the Department of Neurology, University of California, San Francisco
| |
Collapse
|
21
|
Tan GH, Liu YY, Wang L, Li K, Zhang ZQ, Li HF, Yang ZF, Li Y, Li D, Wu MY, Yu CL, Long JJ, Chen RC, Li LX, Yin LP, Liu JW, Cheng XW, Shen Q, Shu YS, Sakimura K, Liao LJ, Wu ZY, Xiong ZQ. PRRT2 deficiency induces paroxysmal kinesigenic dyskinesia by regulating synaptic transmission in cerebellum. Cell Res 2017; 28:90-110. [PMID: 29056747 PMCID: PMC5752836 DOI: 10.1038/cr.2017.128] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/28/2017] [Accepted: 07/26/2017] [Indexed: 01/22/2023] Open
Abstract
Mutations in the proline-rich transmembrane protein 2 (PRRT2) are associated with paroxysmal kinesigenic dyskinesia (PKD) and several other paroxysmal neurological diseases, but the PRRT2 function and pathogenic mechanisms remain largely obscure. Here we show that PRRT2 is a presynaptic protein that interacts with components of the SNARE complex and downregulates its formation. Loss-of-function mutant mice showed PKD-like phenotypes triggered by generalized seizures, hyperthermia, or optogenetic stimulation of the cerebellum. Mutant mice with specific PRRT2 deletion in cerebellar granule cells (GCs) recapitulate the behavioral phenotypes seen in Prrt2-null mice. Furthermore, recording made in cerebellar slices showed that optogenetic stimulation of GCs results in transient elevation followed by suppression of Purkinje cell firing. The anticonvulsant drug carbamazepine used in PKD treatment also relieved PKD-like behaviors in mutant mice. Together, our findings identify PRRT2 as a novel regulator of the SNARE complex and provide a circuit mechanism underlying the PRRT2-related behaviors.
Collapse
Affiliation(s)
- Guo-He Tan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Department of Human Anatomy, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Collaborative Innovation Center of Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yuan-Yuan Liu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lu Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kui Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ze-Qiang Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-Fu Li
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhong-Fei Yang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yang Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming-Yue Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun-Lei Yu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juan-Juan Long
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ren-Chao Chen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Xi Li
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Lu-Ping Yin
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ji-Wei Liu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue-Wen Cheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qi Shen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - You-Sheng Shu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Lu-Jian Liao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Abstract
Headache, especially migraine, has long been associated with epilepsy, based on the common clinical features of these disorders. Both migraine and epilepsy have a genetic predisposition and share common pathophysiological mechanisms including an imbalance between excitatory and inhibitory factors that result in spells of altered brain function and autonomic symptoms. There are well-documented reports on the headache as a sole manifestation of epileptic seizure and headache is commonly associated with as preictal, ictal, and postictal symptoms in epilepsy patients. In addition, migraine and epilepsy are frequently described as highly comorbid conditions and several antiepileptic drugs are used for the patients with migraine as well as epilepsy. In the present review, we briefly discuss the connection between headache and epilepsy in various aspects, including classification, clinical features, epidemiology, genetics, pathophysiology, and treatment.
Collapse
Affiliation(s)
- Dong Wook Kim
- Department of Neurology, Konkuk University School of Medicine, Seoul, Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Wykes RC, Lignani G. Gene therapy and editing: Novel potential treatments for neuronal channelopathies. Neuropharmacology 2017; 132:108-117. [PMID: 28564577 DOI: 10.1016/j.neuropharm.2017.05.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 01/14/2023]
Abstract
Pharmaceutical treatment can be inadequate, non-effective, or intolerable for many people suffering from a neuronal channelopathy. Development of novel treatment options, particularly those with the potential to be curative is warranted. Gene therapy approaches can permit cell-specific modification of neuronal and circuit excitability and have been investigated experimentally as a therapy for numerous neurological disorders, with clinical trials for several neurodegenerative diseases ongoing. Channelopathies can arise from a wide array of gene mutations; however they usually result in periods of aberrant network excitability. Therefore gene therapy strategies based on up or downregulation of genes that modulate neuronal excitability may be effective therapy for a wide range of neuronal channelopathies. As many channelopathies are paroxysmal in nature, optogenetic or chemogenetic approaches may be well suited to treat the symptoms of these diseases. Recent advances in gene-editing technologies such as the CRISPR-Cas9 system could in the future result in entirely novel treatment for a channelopathy by repairing disease-causing channel mutations at the germline level. As the brain may develop and wire abnormally as a consequence of an inherited or de novo channelopathy, the choice of optimal gene therapy or gene editing strategy will depend on the time of intervention (germline, neonatal or adult). This article is part of the Special Issue entitled 'Channelopathies.'
Collapse
Affiliation(s)
- R C Wykes
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, UCL, London, UK.
| | - G Lignani
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, UCL, London, UK.
| |
Collapse
|
24
|
Abu Libdeh A, Talman L, Chambers C, Dhamija R. Clinical Reasoning: A 13-year-old boy with chronic ataxia and developmental delay. Neurology 2017; 88:e116-e121. [DOI: 10.1212/wnl.0000000000003768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
25
|
Kingston WS, Schwedt TJ. The Relationship Between Headaches with Epileptic and Non-epileptic Seizures: a Narrative Review. Curr Pain Headache Rep 2017; 21:17. [DOI: 10.1007/s11916-017-0617-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
26
|
PRRT2: from Paroxysmal Disorders to Regulation of Synaptic Function. Trends Neurosci 2016; 39:668-679. [DOI: 10.1016/j.tins.2016.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022]
|
27
|
Rajapakse T, Buchhalter J. The borderland of migraine and epilepsy in children. Headache 2016; 56:1071-80. [PMID: 27103497 DOI: 10.1111/head.12827] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/08/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To provide a review on the spectrum of migraine-epilepsy disorders in children. BACKGROUND The migraine-epilepsy continuum covers a fascinating array of disorders that share many clinical similarities but also differ fundamentally in pathophysiology. In the pediatric population, its study can be complicated by the young age of those affected and the lack of clear understanding of the neurobiology of these disorders within the developing brain. DISCUSSION This review serves to discuss the borderland of migraine and epilepsy in children. It will focus on epidemiology and comorbidity of the two disorders, possible mechanisms for shared pathophysiology informed by basic and translational science, and an overview of clinical similarities and differences. It will also discuss differentiation of migraine aura from childhood occipital epilepsies. Finally, the review concludes with a discussion of current classification methods for capturing cases on the migraine-epilepsy spectrum and a call for a united approach towards a better definition of this spectrum of disorders. CONCLUSION Recent advances examining the migraine-epilepsy spectrum show clinicopathological similarities between the two disorders in children. Epidemiology demonstrates reciprocally increased incidences of epilepsy in migraineurs and of migraines in children with epilepsy, however, prospective longitudinal in children are currently lacking. Clinically, the two disorders show similarity in preictal, ictal, and postictal phenomena, with close temporal association of the two conditions described by the controversial term of "migralepsy." Basic science research has contributed significant improvements in understanding the generation of both of these episodic neurological conditions, with common links seen at a cellular level involving synaptic glutamate release and the provocation of varying propagation methods including cortical spreading depression in migraine and the paroxysmal depolarizing shift in epilepsy. Despite these significant gains in understanding, improved classification methods are required to identify and further study these interrelated conditions and move towards improved diagnosis and treatment of disorders on the migraine-epilepsy continuum in children.
Collapse
Affiliation(s)
- Thilinie Rajapakse
- Section of Neurology, Alberta Children's Hospital, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeffrey Buchhalter
- Section of Neurology, Alberta Children's Hospital, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Departments of Pediatrics and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta Children's Hospital Research Institute
| |
Collapse
|
28
|
Shamseldin H, Faqeih E, Alasmari A, Zaki M, Gleeson J, Alkuraya F. Mutations in UNC80, Encoding Part of the UNC79-UNC80-NALCN Channel Complex, Cause Autosomal-Recessive Severe Infantile Encephalopathy. Am J Hum Genet 2016; 98:210-5. [PMID: 26708753 DOI: 10.1016/j.ajhg.2015.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/06/2015] [Indexed: 10/22/2022] Open
Abstract
Brain channelopathies represent a growing class of brain disorders that usually result in paroxysmal disorders, although their role in other neurological phenotypes, including the recently described NALCN-related infantile encephalopathy, is increasingly recognized. In three Saudi Arabian families and one Egyptian family all affected by a remarkably similar phenotype (infantile encephalopathy and largely normal brain MRI) to that of NALCN-related infantile encephalopathy, we identified a locus on 2q34 in which whole-exome sequencing revealed three, including two apparently loss-of-function, recessive mutations in UNC80. UNC80 encodes a large protein that is necessary for the stability and function of NALCN and for bridging NALCN to UNC79 to form a functional complex. Our results expand the clinical relevance of the UNC79-UNC80-NALCN channel complex.
Collapse
|
29
|
Spillane J, Kullmann DM, Hanna MG. Genetic neurological channelopathies: molecular genetics and clinical phenotypes. J Neurol Neurosurg Psychiatry 2016; 87:37-48. [PMID: 26558925 PMCID: PMC4717447 DOI: 10.1136/jnnp-2015-311233] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/13/2015] [Indexed: 01/08/2023]
Abstract
Evidence accumulated over recent years has shown that genetic neurological channelopathies can cause many different neurological diseases. Presentations relating to the brain, spinal cord, peripheral nerve or muscle mean that channelopathies can impact on almost any area of neurological practice. Typically, neurological channelopathies are inherited in an autosomal dominant fashion and cause paroxysmal disturbances of neurological function, although the impairment of function can become fixed with time. These disorders are individually rare, but an accurate diagnosis is important as it has genetic counselling and often treatment implications. Furthermore, the study of less common ion channel mutation-related diseases has increased our understanding of pathomechanisms that is relevant to common neurological diseases such as migraine and epilepsy. Here, we review the molecular genetic and clinical features of inherited neurological channelopathies.
Collapse
Affiliation(s)
- J Spillane
- Royal Free Hospital Foundation Trust London, London, UK MRC Centre for Neuromuscular Disease, UCL, London, UK
| | - D M Kullmann
- MRC Centre for Neuromuscular Disease, UCL, London, UK UCL, Institute of Neurology, London, UK
| | - M G Hanna
- MRC Centre for Neuromuscular Disease, UCL, London, UK UCL, Institute of Neurology, London, UK
| |
Collapse
|
30
|
Kaczmarski JA, Corry B. Investigating the size and dynamics of voltage-gated sodium channel fenestrations. Channels (Austin) 2015; 8:264-77. [PMID: 24632677 PMCID: PMC4203756 DOI: 10.4161/chan.28136] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Eukaryotic voltage-gated sodium channels (VGSCs) are essential for the initiation and propagation of action potentials in electrically excitable cells, and are important pharmaceutical targets for the treatment of neurological disorders such as epilepsy, cardiac arrhythmias, and chronic pain. Evidence suggests that small, hydrophobic, VGSC-blocking drugs can gain access to binding residues within the central cavity of these channels by passing through lateral, lipid-filled “fenestrations” which run between the exterior of the protein and its central pore. Here, we use molecular dynamics simulations to investigate how the size and shape of fenestrations change over time in several bacterial VGSC models and a homology model of Nav1.4. We show that over the course of the simulations, the size of the fenestrations is primarily influenced by rapid protein motions, such as amino acid side-chain rotation, and highlight that differences between fenestration bottleneck-contributing residues are the primary cause of variations in fenestration size between the 6 bacterial models. In the eukaryotic channel model, 2 fenestrations are wide, but 2 are narrow due to differences in the amino acid sequence in the 4 domains. Lipid molecules are found to influence the size of the fenestrations by protruding acylchains into the fenestrations and displacing amino acid side-chains. Together, the results suggest that fenestrations provide viable pathways for small, flexible, hydrophobic drugs.
Collapse
|
31
|
Abnormal excitability and episodic low-frequency oscillations in the cerebral cortex of the tottering mouse. J Neurosci 2015; 35:5664-79. [PMID: 25855180 DOI: 10.1523/jneurosci.3107-14.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The Ca(2+) channelopathies caused by mutations of the CACNA1A gene that encodes the pore-forming subunit of the human Cav2.1 (P/Q-type) voltage-gated Ca(2+) channel include episodic ataxia type 2 (EA2). Although, in EA2 the emphasis has been on cerebellar dysfunction, patients also exhibit episodic, nonmotoric abnormalities involving the cerebral cortex. This study demonstrates episodic, low-frequency oscillations (LFOs) throughout the cerebral cortex of tottering (tg/tg) mice, a widely used model of EA2. Ranging between 0.035 and 0.11 Hz, the LFOs in tg/tg mice can spontaneously develop very high power, referred to as a high-power state. The LFOs in tg/tg mice are mediated in part by neuronal activity as tetrodotoxin decreases the oscillations and cortical neuron discharge contain the same low frequencies. The high-power state involves compensatory mechanisms because acutely decreasing P/Q-type Ca(2+) channel function in either wild-type (WT) or tg/tg mice does not induce the high-power state. In contrast, blocking l-type Ca(2+) channels, known to be upregulated in tg/tg mice, reduces the high-power state. Intriguingly, basal excitatory glutamatergic neurotransmission constrains the high-power state because blocking ionotropic or metabotropic glutamate receptors results in high-power LFOs in tg/tg but not WT mice. The high-power LFOs are decreased markedly by acetazolamide and 4-aminopyridine, the primary treatments for EA2, suggesting disease relevance. Together, these results demonstrate that the high-power LFOs in the tg/tg cerebral cortex represent a highly abnormal excitability state that may underlie noncerebellar symptoms that characterize CACNA1A mutations.
Collapse
|
32
|
Protein mutated in paroxysmal dyskinesia interacts with the active zone protein RIM and suppresses synaptic vesicle exocytosis. Proc Natl Acad Sci U S A 2015; 112:2935-41. [PMID: 25730884 DOI: 10.1073/pnas.1501364112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Paroxysmal nonkinesigenic dyskinesia (PNKD) is an autosomal dominant episodic movement disorder precipitated by coffee, alcohol, and stress. We previously identified the causative gene but the function of the encoded protein remains unknown. We also generated a PNKD mouse model that revealed dysregulated dopamine signaling in vivo. Here, we show that PNKD interacts with synaptic active zone proteins Rab3-interacting molecule (RIM)1 and RIM2, localizes to synapses, and modulates neurotransmitter release. Overexpressed PNKD protein suppresses release, and mutant PNKD protein is less effective than wild-type at inhibiting exocytosis. In PNKD KO mice, RIM1/2 protein levels are reduced and synaptic strength is impaired. Thus, PNKD is a novel synaptic protein with a regulatory role in neurotransmitter release.
Collapse
|
33
|
Affiliation(s)
- Louis J Ptáček
- Department of Neurology, Howard Hughes Medical Institute, University of California, San Francisco, California 94158-2324;
| |
Collapse
|
34
|
Kim GE, Kronengold J, Barcia G, Quraishi IH, Martin HC, Blair E, Taylor JC, Dulac O, Colleaux L, Nabbout R, Kaczmarek LK. Human slack potassium channel mutations increase positive cooperativity between individual channels. Cell Rep 2014; 9:1661-1672. [PMID: 25482562 DOI: 10.1016/j.celrep.2014.11.015] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 10/06/2014] [Accepted: 11/10/2014] [Indexed: 11/24/2022] Open
Abstract
Disease-causing mutations in ion channels generally alter intrinsic gating properties such as activation, inactivation, and voltage dependence. We examined nine different mutations of the KCNT1 (Slack) Na(+)-activated K(+) channel that give rise to three distinct forms of epilepsy. All produced many-fold increases in current amplitude compared to the wild-type channel. This could not be accounted for by increases in the intrinsic open probability of individual channels. Rather, greatly increased opening was a consequence of cooperative interactions between multiple channels in a patch. The degree of cooperative gating was much greater for all of the mutant channels than for the wild-type channel, and could explain increases in current even in a mutant with reduced unitary conductance. We also found that the same mutation gave rise to different forms of epilepsy in different individuals. Our findings indicate that a major consequence of these mutations is to alter channel-channel interactions.
Collapse
Affiliation(s)
- Grace E Kim
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Jack Kronengold
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA
| | - Giulia Barcia
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Imran H Quraishi
- Comprehensive Epilepsy Center, Department of Neurology, Yale University, New Haven, CT 06520, USA
| | - Hilary C Martin
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Edward Blair
- Oxford University Hospitals Trust, Oxford OX3 9DU, UK
| | - Jenny C Taylor
- Oxford Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Olivier Dulac
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Laurence Colleaux
- INSERM U781, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Rima Nabbout
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
35
|
Rahman T, Cai X, Brailoiu GC, Abood ME, Brailoiu E, Patel S. Two-pore channels provide insight into the evolution of voltage-gated Ca2+ and Na+ channels. Sci Signal 2014; 7:ra109. [PMID: 25406377 PMCID: PMC4327855 DOI: 10.1126/scisignal.2005450] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Four-domain voltage-gated Ca(2+) and Na(+) channels (CaV, NaV) underpin nervous system function and likely emerged upon intragenic duplication of a primordial two-domain precursor. To investigate if two-pore channels (TPCs) may represent an intermediate in this evolutionary transition, we performed molecular docking simulations with a homology model of TPC1, which suggested that the pore region could bind antagonists of CaV or NaV. CaV or NaV antagonists blocked NAADP (nicotinic acid adenine dinucleotide phosphate)-evoked Ca(2+) signals in sea urchin egg preparations and in intact cells that overexpressed TPC1. By sequence analysis and inspection of the model, we predicted a noncanonical selectivity filter in animal TPCs in which the carbonyl groups of conserved asparagine residues are positioned to coordinate cations. In contrast, a distinct clade of TPCs [TPCR (for TPC-related)] in several unicellular species had ion selectivity filters with acidic residues more akin to CaV. TPCRs were predicted to interact strongly with CaV antagonists. Our data suggest that acquisition of a "blueprint" pharmacological profile and changes in ion selectivity within four-domain voltage-gated ion channels may have predated intragenic duplication of an ancient two-domain ancestor.
Collapse
Affiliation(s)
- Taufiq Rahman
- Department of Pharmacology, Cambridge University, Cambridge CB2 1PD, UK.
| | - Xinjiang Cai
- Department of Cell Developmental Biology, University College London, London WC1E 6BT, UK
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University School of Pharmacy, Philadelphia, PA 19107, USA
| | - Mary E Abood
- Department of Anatomy and Cell Biology and Center for Substance Abuse Research, Temple University, Philadelphia, PA 19140, USA
| | - Eugen Brailoiu
- Department of Pharmacology and Center for Substance Abuse Research, Temple University, Philadelphia, PA 19140, USA
| | - Sandip Patel
- Department of Cell Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
36
|
Abstract
As noted in the separate introduction to this special topic section, episodic and electrical disorders can appear quite different clinically and yet share many overlapping features, including attack precipitants, therapeutic responses, natural history, and the types of genes that cause many of the genetic forms (i.e., ion channel genes). Thus, as we mapped and attempted to clone genes causing other episodic disorders, ion channels were always outstanding candidates when they mapped to the critical region of linkage in such a family. However, some of these disorders do not result from mutations in channels. This realization has opened up large and exciting new areas for the pathogenesis of these disorders. In some cases, the mutations occur in genes of unknown function or without understanding of molecular pathogenesis. Recently, emerging insights into a fascinating group of episodic movement disorders, the paroxysmal dyskinesias, and study of the causative genes and proteins are leading to the emerging concept of episodic electric disorders resulting from synaptic dysfunction. Much work remains to be done, but the field is evolving rapidly. As it does, we have come to realize that the molecular pathogenesis of electrical and episodic disorders is more complex than a scenario in which such disorders are simply due to mutations in the primary determinants of membrane excitability (channels).
Collapse
|
37
|
Lim CH, Zain SM, Reynolds GP, Zain MA, Roffeei SN, Zainal NZ, Kanagasundram S, Mohamed Z. Genetic association of LMAN2L gene in schizophrenia and bipolar disorder and its interaction with ANK3 gene polymorphism. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:157-62. [PMID: 24914473 DOI: 10.1016/j.pnpbp.2014.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/19/2014] [Accepted: 05/29/2014] [Indexed: 12/22/2022]
Abstract
Recent studies have shown that bipolar disorder (BPD) and schizophrenia (SZ) share some common genetic risk factors. This study aimed to examine the association between candidate single nucleotide polymorphisms (SNPs) identified from genome-wide association studies (GWAS) and risk of BPD and SZ. A total of 715 patients (244 BPD and 471 SZ) and 593 controls were genotyped using the Sequenom MassARRAY platform. We showed a positive association between LMAN2L (rs6746896) and risk of both BPD and SZ in a pooled population (P-value=0.001 and 0.009, respectively). Following stratification by ethnicity, variants of the ANK3 gene (rs1938516 and rs10994336) were found to be associated with BPD in Malays (P-value=0.001 and 0.006, respectively). Furthermore, an association exists between another variant of LMAN2L (rs2271893) and SZ in the Malay and Indian ethnic groups (P-value=0.003 and 0.002, respectively). Gene-gene interaction analysis revealed a significant interaction between the ANK3 and LMAN2L genes (empirical P=0.0107). Significant differences were shown between patients and controls for two haplotype frequencies of LMAN2L: GA (P=0.015 and P=0.010, for BPD and SZ, respectively) and GG (P=0.013 for BPD). Our study showed a significant association between LMAN2L and risk of both BPD and SZ.
Collapse
Affiliation(s)
- Chor Hong Lim
- The Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Shamsul Mohd Zain
- The Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Gavin P Reynolds
- Biomedical Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield S11WB, UK
| | - Mohd Aizat Zain
- The Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Siti Norsyuhada Roffeei
- The Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nor Zuraida Zainal
- Department of Psychological Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sharmilla Kanagasundram
- Department of Psychological Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- The Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Mao CY, Shi CH, Song B, Wu J, Ji Y, Qin J, Li YS, Wang JJ, Shang DD, Sun SL, Xu YM. Genotype–phenotype correlation in a cohort of paroxysmal kinesigenic dyskinesia cases. J Neurol Sci 2014; 340:91-3. [DOI: 10.1016/j.jns.2014.02.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/07/2014] [Accepted: 02/25/2014] [Indexed: 11/29/2022]
|
39
|
Frank CA. How voltage-gated calcium channels gate forms of homeostatic synaptic plasticity. Front Cell Neurosci 2014; 8:40. [PMID: 24592212 PMCID: PMC3924756 DOI: 10.3389/fncel.2014.00040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/28/2014] [Indexed: 01/15/2023] Open
Abstract
Throughout life, animals face a variety of challenges such as developmental growth, the presence of toxins, or changes in temperature. Neuronal circuits and synapses respond to challenges by executing an array of neuroplasticity paradigms. Some paradigms allow neurons to up- or downregulate activity outputs, while countervailing ones ensure that outputs remain within appropriate physiological ranges. A growing body of evidence suggests that homeostatic synaptic plasticity (HSP) is critical in the latter case. Voltage-gated calcium channels gate forms of HSP. Presynaptically, the aggregate data show that when synapse activity is weakened, homeostatic signaling systems can act to correct impairments, in part by increasing calcium influx through presynaptic CaV2-type channels. Increased calcium influx is often accompanied by parallel increases in the size of active zones and the size of the readily releasable pool of presynaptic vesicles. These changes coincide with homeostatic enhancements of neurotransmitter release. Postsynaptically, there is a great deal of evidence that reduced network activity and loss of calcium influx through CaV1-type calcium channels also results in adaptive homeostatic signaling. Some adaptations drive presynaptic enhancements of vesicle pool size and turnover rate via retrograde signaling, as well as de novo insertion of postsynaptic neurotransmitter receptors. Enhanced calcium influx through CaV1 after network activation or single cell stimulation can elicit the opposite response-homeostatic depression via removal of excitatory receptors. There exist intriguing links between HSP and calcium channelopathies-such as forms of epilepsy, migraine, ataxia, and myasthenia. The episodic nature of some of these disorders suggests alternating periods of stable and unstable function. Uncovering information about how calcium channels are regulated in the context of HSP could be relevant toward understanding these and other disorders.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| |
Collapse
|
40
|
Martinez-Martinez P, Molenaar PC, Losen M, Stevens J, Baets MHD, Szoke A, Honnorat J, Tamouza R, Leboyer M, Os JV, Rutten BPF. Autoantibodies to neurotransmitter receptors and ion channels: from neuromuscular to neuropsychiatric disorders. Front Genet 2013; 4:181. [PMID: 24065983 PMCID: PMC3778371 DOI: 10.3389/fgene.2013.00181] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 08/27/2013] [Indexed: 01/17/2023] Open
Abstract
Changes of voltage-gated ion channels and ligand-gated receptor channels caused by mutation or autoimmune attack are the cause of so-called channelopathies in the central and peripheral nervous system. We present the pathophysiology of channelopathies of the neuromuscular junction in terms of loss-of-function and gain-of-function principles. Autoantibodies generally have reduced access to the central nervous system, but in some cases this is enough to cause disease. A review is provided of recent findings implicating autoantibodies against ligand-activated receptor channels and potassium channels in psychiatric and neurological disorders, including schizophrenia and limbic encephalitis. The emergence of channelopathy-related neuropsychiatric disorders has implications for research and practice.
Collapse
Affiliation(s)
- Pilar Martinez-Martinez
- Department of Psychiatry and Psychology, School for Mental Health and Neuroscience, Maastricht University Maastricht, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Waszkielewicz AM, Gunia A, Szkaradek N, Słoczyńska K, Krupińska S, Marona H. Ion channels as drug targets in central nervous system disorders. Curr Med Chem 2013; 20:1241-85. [PMID: 23409712 PMCID: PMC3706965 DOI: 10.2174/0929867311320100005] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 01/14/2013] [Accepted: 01/18/2013] [Indexed: 12/27/2022]
Abstract
Ion channel targeted drugs have always been related with either the central nervous system (CNS), the peripheral nervous system, or the cardiovascular system. Within the CNS, basic indications of drugs are: sleep disorders, anxiety, epilepsy, pain, etc. However, traditional channel blockers have multiple adverse events, mainly due to low specificity of mechanism of action. Lately, novel ion channel subtypes have been discovered, which gives premises to drug discovery process led towards specific channel subtypes. An example is Na(+) channels, whose subtypes 1.3 and 1.7-1.9 are responsible for pain, and 1.1 and 1.2 - for epilepsy. Moreover, new drug candidates have been recognized. This review is focusing on ion channels subtypes, which play a significant role in current drug discovery and development process. The knowledge on channel subtypes has developed rapidly, giving new nomenclatures of ion channels. For example, Ca(2+)s channels are not any more divided to T, L, N, P/Q, and R, but they are described as Ca(v)1.1-Ca(v)3.3, with even newer nomenclature α1A-α1I and α1S. Moreover, new channels such as P2X1-P2X7, as well as TRPA1-TRPV1 have been discovered, giving premises for new types of analgesic drugs.
Collapse
Affiliation(s)
- A M Waszkielewicz
- Department of Bioorganic Chemistry, Chair of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Many neurologic diseases cause discrete episodic impairment in contrast with progressive deterioration. The symptoms of these episodic disorders exhibit striking variety. Herein we review what is known of the phenotypes, genetics, and pathophysiology of episodic neurologic disorders. Of these, most are genetically complex, with unknown or polygenic inheritance. In contrast, a fascinating panoply of episodic disorders exhibit Mendelian inheritance. We classify episodic Mendelian disorders according to the primary neuroanatomical location affected: skeletal muscle, cardiac muscle, neuromuscular junction, peripheral nerve, or central nervous system (CNS). Most known Mendelian mutations alter genes that encode membrane-bound ion channels. These mutations cause ion channel dysfunction, which ultimately leads to altered membrane excitability as manifested by episodic disease. Other Mendelian disease genes encode proteins essential for ion channel trafficking or stability. These observations have cemented the channelopathy paradigm, in which episodic disorders are conceptualized as disorders of ion channels. However, we expand on this paradigm to propose that dysfunction at the synaptic and neuronal circuit levels may underlie some episodic neurologic entities.
Collapse
Affiliation(s)
- Jonathan F Russell
- Department of Neurology, Howard Hughes Medical Institute, School of Medicine, University of California-San Francisco, CA 94158, USA.
| | | | | |
Collapse
|
43
|
Judy JT, Zandi PP. A review of potassium channels in bipolar disorder. Front Genet 2013; 4:105. [PMID: 23781230 PMCID: PMC3678088 DOI: 10.3389/fgene.2013.00105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/22/2013] [Indexed: 12/11/2022] Open
Abstract
Although bipolar disorder (BP) is one of the most heritable psychiatric conditions, susceptibility genes for the disorder have yet to be conclusively identified. It is likely that variants in multiple genes across multiple pathways contribute to the genotype–phenotype relationship in the affected population. Recent evidence from genome-wide association studies implicates an entire class of genes related to the structure and regulation of ion channels, suggesting that the etiology of BP may arise from channelopathies. In this review, we examine the evidence for this hypothesis, with a focus on the potential role of voltage-gated potassium channels. We consider evidence from genetic and expression studies, and discuss the potential underlying biology. We consider animal models and treatment implications of the involvement of potassium ion channelopathy in BP. Finally, we explore intriguing parallels between BP and epilepsy, the signature channelopathy of the central nervous system.
Collapse
Affiliation(s)
- Jennifer T Judy
- Department of Psychiatry, Johns Hopkins School of Medicine Baltimore, MD, USA
| | | |
Collapse
|
44
|
Rodriguez-Sainz A, Pinedo-Brochado A, Sánchez-Menoyo JL, Ruiz-Ojeda J, Escalza-Cortina I, Garcia-Monco JC. Migraine, Stroke and Epilepsy: Underlying and Interrelated Causes, Diagnosis and Treatment. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2013; 15:322-34. [DOI: 10.1007/s11936-013-0236-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Kuo SH, Xie T, Fahn S. Anticholinergic responsive cyclic myoclonus. Mov Disord 2013; 28:401-2. [PMID: 23404345 DOI: 10.1002/mds.25284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/11/2012] [Accepted: 10/21/2012] [Indexed: 11/08/2022] Open
|
46
|
Gardiner AR, Bhatia KP, Stamelou M, Dale RC, Kurian MA, Schneider SA, Wali GM, Counihan T, Schapira AH, Spacey SD, Valente EM, Silveira-Moriyama L, Teive HAG, Raskin S, Sander JW, Lees A, Warner T, Kullmann DM, Wood NW, Hanna M, Houlden H. PRRT2 gene mutations: from paroxysmal dyskinesia to episodic ataxia and hemiplegic migraine. Neurology 2012; 79:2115-21. [PMID: 23077024 DOI: 10.1212/wnl.0b013e3182752c5a] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE The proline-rich transmembrane protein (PRRT2) gene was recently identified using exome sequencing as the cause of autosomal dominant paroxysmal kinesigenic dyskinesia (PKD) with or without infantile convulsions (IC) (PKD/IC syndrome). Episodic neurologic disorders, such as epilepsy, migraine, and paroxysmal movement disorders, often coexist and are thought to have a shared channel-related etiology. To investigate further the frequency, spectrum, and phenotype of PRRT2 mutations, we analyzed this gene in 3 large series of episodic neurologic disorders with PKD/IC, episodic ataxia (EA), and hemiplegic migraine (HM). METHODS The PRRT2 gene was sequenced in 58 family probands/sporadic individuals with PKD/IC, 182 with EA, 128 with HM, and 475 UK and 96 Asian controls. RESULTS PRRT2 genetic mutations were identified in 28 out of 58 individuals with PKD/IC (48%), 1/182 individuals with EA, and 1/128 individuals with HM. A number of loss-of-function and coding missense mutations were identified; the most common mutation found was the p.R217Pfs*8 insertion. Males were more frequently affected than females (ratio 52:32). There was a high proportion of PRRT2 mutations found in families and sporadic cases with PKD associated with migraine or HM (10 out of 28). One family had EA with HM and another large family had typical HM alone. CONCLUSIONS This work expands the phenotype of mutations in the PRRT2 gene to include the frequent occurrence of migraine and HM with PKD/IC, and the association of mutations with EA and HM and with familial HM alone. We have also extended the PRRT2 mutation type and frequency in PKD and other episodic neurologic disorders.
Collapse
Affiliation(s)
- Alice R Gardiner
- Department of Molecular Neuroscience and Reta Lila Weston Laboratories, MRC Centre for Neuromuscular Diseases, Children's Hospital at Westmead, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Strength-duration time constant in peripheral nerve: no abnormality in multiple sclerosis. Mult Scler Int 2012; 2012:390157. [PMID: 22645684 PMCID: PMC3356942 DOI: 10.1155/2012/390157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 03/13/2012] [Accepted: 03/20/2012] [Indexed: 11/23/2022] Open
Abstract
Objectives. To investigate the properties of the strength-duration time constant (SDTC) in multiple sclerosis (MS) patients. Methods. The SDTC and rheobase in 16 MS patients and 19 healthy controls were obtained following stimulation of the right median nerve at the wrist. Results. SDTC and rheobase values were 408.3 ± 60.0 μs and 4.0 ± 1.8 mA in MS patients, versus 408.0 ± 62.4 μs and 3.8 ± 2.1 mA in controls. The differences were not significant in SDTC or rheobase values between the patients and controls (P = 0.988 for SDTC and P = 0.722 for rheobase). Conclusion. Our study showed no abnormality in relapsing remitting MS patients in terms of SDTC, which gives some indirect information about peripheral Na+ channel function. This may indicate that alterations in the Na+ channel pattern in central nervous system (CNS) couldnot be shown in the peripheral nervous system (PNS) in the MS patients by SDTC. The opinion that MS can be a kind of channelopathy might be proven by performing other axonal excitability tests or SDTC in progressive forms of MS.
Collapse
|
48
|
Lee HY, Huang Y, Bruneau N, Roll P, Roberson EDO, Hermann M, Quinn E, Maas J, Edwards R, Ashizawa T, Baykan B, Bhatia K, Bressman S, Bruno MK, Brunt ER, Caraballo R, Echenne B, Fejerman N, Frucht S, Gurnett CA, Hirsch E, Houlden H, Jankovic J, Lee WL, Lynch DR, Mohammed S, Müller U, Nespeca MP, Renner D, Rochette J, Rudolf G, Saiki S, Soong BW, Swoboda KJ, Tucker S, Wood N, Hanna M, Bowcock AM, Szepetowski P, Fu YH, Ptáček LJ. Mutations in the gene PRRT2 cause paroxysmal kinesigenic dyskinesia with infantile convulsions. Cell Rep 2011; 1:2-12. [PMID: 22832103 DOI: 10.1016/j.celrep.2011.11.001] [Citation(s) in RCA: 199] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/21/2011] [Accepted: 11/07/2011] [Indexed: 11/25/2022] Open
Abstract
Paroxysmal kinesigenic dyskinesia with infantile convulsions (PKD/IC) is an episodic movement disorder with autosomal-dominant inheritance and high penetrance, but the causative genetic mutation is unknown. We have now identified four truncating mutations involving the gene PRRT2 in the vast majority (24/25) of well-characterized families with PKD/IC. PRRT2 truncating mutations were also detected in 28 of 78 additional families. PRRT2 encodes a proline-rich transmembrane protein of unknown function that has been reported to interact with the t-SNARE, SNAP25. PRRT2 localizes to axons but not to dendritic processes in primary neuronal culture, and mutants associated with PKD/IC lead to dramatically reduced PRRT2 levels, leading ultimately to neuronal hyperexcitability that manifests in vivo as PKD/IC.
Collapse
Affiliation(s)
- Hsien-Yang Lee
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Aimon S, Manzi J, Schmidt D, Poveda Larrosa JA, Bassereau P, Toombes GES. Functional reconstitution of a voltage-gated potassium channel in giant unilamellar vesicles. PLoS One 2011; 6:e25529. [PMID: 21998666 PMCID: PMC3188570 DOI: 10.1371/journal.pone.0025529] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 09/05/2011] [Indexed: 11/19/2022] Open
Abstract
Voltage-gated ion channels are key players in cellular excitability. Recent studies suggest that their behavior can depend strongly on the membrane lipid composition and physical state. In vivo studies of membrane/channel and channel/channel interactions are challenging as membrane properties are actively regulated in living cells, and are difficult to control in experimental settings. We developed a method to reconstitute functional voltage-gated ion channels into cell-sized Giant Unilamellar Vesicles (GUVs) in which membrane composition, tension and geometry can be controlled. First, a voltage-gated potassium channel, KvAP, was purified, fluorescently labeled and reconstituted into small proteoliposomes. Small proteoliposomes were then converted into GUVs via electroformation. GUVs could be formed using different lipid compositions and buffers containing low (5 mM) or near-physiological (100 mM) salt concentrations. Protein incorporation into GUVs was characterized with quantitative confocal microscopy, and the protein density of GUVs was comparable to the small proteoliposomes from which they were formed. Furthermore, patch-clamp measurements confirmed that the reconstituted channels retained potassium selectivity and voltage-gated activation. GUVs containing functional voltage-gated ion channels will allow the study of channel activity, distribution and diffusion while controlling membrane state, and should prove a powerful tool for understanding how the membrane modulates cellular excitability.
Collapse
Affiliation(s)
- Sophie Aimon
- Unité Mixte de Recherche (UMR) 168, Physico-Chimie Curie, Centre National de la Recherche Scientifique (CNRS), Institut Curie, Centre de Recherche, Université Pierre et Marie Curie, Paris, France
| | - John Manzi
- Unité Mixte de Recherche (UMR) 168, Physico-Chimie Curie, Centre National de la Recherche Scientifique (CNRS), Institut Curie, Centre de Recherche, Université Pierre et Marie Curie, Paris, France
| | - Daniel Schmidt
- Howard Hughes Medical Institute, Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, New York, United States of America
| | | | - Patricia Bassereau
- Unité Mixte de Recherche (UMR) 168, Physico-Chimie Curie, Centre National de la Recherche Scientifique (CNRS), Institut Curie, Centre de Recherche, Université Pierre et Marie Curie, Paris, France
- * E-mail:
| | - Gilman E. S. Toombes
- Unité Mixte de Recherche (UMR) 168, Physico-Chimie Curie, Centre National de la Recherche Scientifique (CNRS), Institut Curie, Centre de Recherche, Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
50
|
Diverse roles for auxiliary subunits in phosphorylation-dependent regulation of mammalian brain voltage-gated potassium channels. Pflugers Arch 2011; 462:631-43. [PMID: 21822597 DOI: 10.1007/s00424-011-1004-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 07/22/2011] [Accepted: 07/22/2011] [Indexed: 10/17/2022]
Abstract
Voltage-gated ion channels are a diverse family of signaling proteins that mediate rapid electrical signaling events. Among these, voltage-gated potassium or Kv channels are the most diverse partly due to the large number of principal (or α) subunits and auxiliary subunits that can assemble in different combinations to generate Kv channel complexes with distinct structures and functions. The diversity of Kv channels underlies much of the variability in the active properties between different mammalian central neurons and the dynamic changes that lead to experience-dependent plasticity in intrinsic excitability. Recent studies have revealed that Kv channel α subunits and auxiliary subunits are extensively phosphorylated, contributing to additional structural and functional diversity. Here, we highlight recent studies that show that auxiliary subunits exert some of their profound effects on dendritic Kv4 and axonal Kv1 channels through phosphorylation-dependent mechanisms, either due to phosphorylation on the auxiliary subunit itself or by influencing the extent and/or impact of α subunit phosphorylation. The complex effects of auxiliary subunits and phosphorylation provide a potent mechanism to generate additional diversity in the structure and function of Kv4 and Kv1 channels, as well as allowing for dynamic reversible regulation of these important ion channels.
Collapse
|