1
|
Waris A, Siraj M, Khan A, Lin J, Asim M, Alhumaydh FA. A Comprehensive Overview of the Current Status and Advancements in Various Treatment Strategies against Epilepsy. ACS Pharmacol Transl Sci 2024; 7:3729-3757. [PMID: 39698272 PMCID: PMC11650742 DOI: 10.1021/acsptsci.4c00494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Epilepsy affects more than 70 million individuals of all ages worldwide and remains one of the most severe chronic noncommunicable neurological diseases globally. Several neurotransmitters, membrane protein channels, receptors, enzymes, and, more recently noted, various pathways, such as inflammatory and mTORC complexes, play significant roles in the initiation and propagation of seizures. Over the past two decades, significant developments have been made in the diagnosis and treatment of epilepsy. Various pharmacological drugs with diverse mechanisms of action and other treatment options have been developed to control seizures and treat epilepsy. These options include surgical treatment, nanomedicine, gene therapy, natural products, nervous stimulation, a ketogenic diet, gut microbiota, etc., which are in various developmental stages. Despite a plethora of drugs and other treatment options, one-third of affected individuals are resistant to current medications, while the majority of approved drugs have severe side effects, and significant changes can occur, such as pharmacoresistance, effects on cognition, long-term problems, drug interactions, risks of poor adherence, specific effects for certain medications, and psychological complications. Therefore, the development of new drugs and other treatment options that have no or minimal adverse effects is needed to combat this deadly disease. In this Review, we comprehensively summarize and explain all of the treatment options that have been approved or are in developmental stages for epilepsy as well as their status in clinical trials and advancements.
Collapse
Affiliation(s)
- Abdul Waris
- Department
of Biomedical Science, City University of
Hong Kong, 999077 Hong Kong SAR
| | - Muhammad Siraj
- Department
of Biotechnology, Jeonbuk National University−Iksan
Campus, Jeonju 54896, South Korea
| | - Ayyaz Khan
- Department
of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, South Korea
| | - Junyu Lin
- Department
of Neuroscience, City University of Hong
Kong, 999077 Hong Kong SAR
| | - Muhammad Asim
- Department
of Neuroscience, City University of Hong
Kong, 999077 Hong Kong SAR
| | - Fahad A. Alhumaydh
- Department
of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
2
|
Zhang Y, Wu J, Zheng Y, Xu Y, Yu Z, Ping Y. Voltage Gated Ion Channels and Sleep. J Membr Biol 2024; 257:269-280. [PMID: 39354150 DOI: 10.1007/s00232-024-00325-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Ion channels are integral components of the nervous system, playing a pivotal role in shaping membrane potential, neuronal excitability, synaptic transmission and plasticity. Dysfunction in these channels, such as improper expression or localization, can lead to irregular neuronal excitability and synaptic communication, which may manifest as various behavioral abnormalities, including disrupted rest-activity cycles. Research has highlighted the significant impact of voltage gated ion channels on sleep parameters, influencing sleep latency, duration and waveforms. Furthermore, these ion channels have been implicated in the vulnerability to, and the pathogenesis of, several neurological and psychiatric disorders, including epilepsy, autism, schizophrenia, and Alzheimer's disease (AD). In this comprehensive review, we aim to provide a summary of the regulatory role of three predominant types of voltage-gated ion channels-calcium (Ca2+), sodium (Na+), and potassium (K+)-in sleep across species, from flies to mammals. We will also discuss the association of sleep disorders with various human diseases that may arise from the dysfunction of these ion channels, thereby underscoring the potential therapeutic benefits of targeting specific ion channel subtypes for sleep disturbance treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiawen Wu
- Faculty of Brain Sciences, University College London, London, UK
| | - Yuxian Zheng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yangkun Xu
- Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Ziqi Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
3
|
Li Z, Wu Q, Yan N. A structural atlas of druggable sites on Na v channels. Channels (Austin) 2024; 18:2287832. [PMID: 38033122 PMCID: PMC10732651 DOI: 10.1080/19336950.2023.2287832] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Voltage-gated sodium (Nav) channels govern membrane excitability by initiating and propagating action potentials. Consistent with their physiological significance, dysfunction, or mutations in these channels are associated with various channelopathies. Nav channels are thereby major targets for various clinical and investigational drugs. In addition, a large number of natural toxins, both small molecules and peptides, can bind to Nav channels and modulate their functions. Technological breakthrough in cryo-electron microscopy (cryo-EM) has enabled the determination of high-resolution structures of eukaryotic and eventually human Nav channels, alone or in complex with auxiliary subunits, toxins, and drugs. These studies have not only advanced our comprehension of channel architecture and working mechanisms but also afforded unprecedented clarity to the molecular basis for the binding and mechanism of action (MOA) of prototypical drugs and toxins. In this review, we will provide an overview of the recent advances in structural pharmacology of Nav channels, encompassing the structural map for ligand binding on Nav channels. These findings have established a vital groundwork for future drug development.
Collapse
Affiliation(s)
- Zhangqiang Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiurong Wu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong Province, China
| |
Collapse
|
4
|
Zhang X, Qiao K, Cui R, Xu M, Cai S, Huang Q, Liu Z. Tetrodotoxin: The State-of-the-Art Progress in Characterization, Detection, Biosynthesis, and Transport Enrichment. Mar Drugs 2024; 22:531. [PMID: 39728106 DOI: 10.3390/md22120531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Tetrodotoxin (TTX) is a neurotoxin that binds to sodium channels and blocks sodium conduction. Importantly, TTX has been increasingly detected in edible aquatic organisms. Because of this and the lack of specific antidotes, TTX poisoning is now a major threat to public health. However, it is of note that ultra-low dose TTX is an excellent analgesic with great medicinal value. These contradictory effects highlight the need for further research to elucidate the impacts and functional mechanisms of TTX. This review summarizes the latest research progress in relation to TTX sources, analogs, mechanisms of action, detection methods, poisoning symptoms, therapeutic options, biosynthesis pathways, and mechanisms of transport and accumulation in pufferfish. This review also provides a theoretical basis for reducing the poisoning risks associated with TTX and for establishing an effective system for its use and management to ensure the safety of fisheries and human health.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Kun Qiao
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen 361021, China
| | - Ruimin Cui
- College of Food Science and Technology, Zhejiang Ocean University, Zhoushan 316022, China
| | - Min Xu
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen 361021, China
| | - Shuilin Cai
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen 361021, China
| | - Qilin Huang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhiyu Liu
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen 361021, China
| |
Collapse
|
5
|
Iyer SH, Simeone KA. Salty Battle to Sudden Death: Young vs. Old, Brain vs. Heart. Epilepsy Curr 2024; 24:437-439. [PMID: 39540134 PMCID: PMC11556351 DOI: 10.1177/15357597241289672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Cardiac-Specific Deletion of Scn8a Mitigates Dravet Syndrome-Associated Sudden Death in Adults King DR, Demirtas M, Tarasov M, Struckman HL, Meng X, Nassal D, Moise N, Miller A, Min D, Soltisz AM, Anne MNK, Alves DPA, Wagnon JL, Weinberg SH, Hund TJ, Veeraraghavan R, Radwański PB. JACC Clin. Electrophysiol . 2024;10(5):829–842. doi:10.1016/j.jacep.2024.01.003. Sudden unexpected death in epilepsy (SUDEP) is a fatal complication experienced by otherwise healthy epilepsy patients. Dravet syndrome (DS) is an inherited epileptic disorder resulting from loss-of-function of the voltage-gated sodium channel, NaV 1.1, and is associated with particularly high SUDEP risk. Evidence is mounting that NaVs abundant in the brain also occur in the heart, suggesting that the very molecular mechanisms underlying epilepsy could also precipitate cardiac arrhythmias and sudden death. Despite marked reduction of NaV 1.1 functional expression in DS, pathogenic late sodium current (INa,L) is paradoxically increased in DS hearts. However, the mechanisms by which DS directly impacts the heart to promote sudden death remain unclear. In this study, the authors sought to provide evidence implicating remodeling of Na+- and Ca2+-handling machinery, including NaV 1.6 and Na+/Ca2+ exchanger (NCX) within transverse (T)-tubules in DS-associated arrhythmias. The authors undertook scanning ion conductance microscopy (SICM)-guided patch clamp, super-resolution microscopy, confocal Ca2+ imaging, and in vivo electrocardiography studies in Scn1a haploinsufficient murine model of DS. DS promotes INa,L in T-tubular nanodomains, but not in other subcellular regions. Consistent with increased NaV activity in these regions, super-resolution microscopy revealed increased NaV 1.6 density near Ca2+ release channels, the ryanodine receptors (RyR2), and NCX in DS relative to WT hearts. The resulting INa,L in these regions promoted aberrant Ca2+ release, leading to ventricular arrhythmias in vivo. Cardiac-specific deletion of NaV 1.6 protects adult DS mice from increased T-tubular late NaV activity and the resulting arrhythmias, as well as sudden death. These data demonstrate that NaV 1.6 undergoes remodeling within T-tubules of adult DS hearts serving as a substrate for Ca2+-mediated cardiac arrhythmias and may be a druggable target for the prevention of SUDEP in adult DS subjects. Molecular and Cellular Context Influences SCN8A Variant Function Vanoye CG, Abramova TV, DeKeyser J-M, Ghabra NF, Oudin MJ, Burge CB, Helbig I, Thompson CH, George AL. JCI Insight 2024; 9(12). doi:10.1172/jci.insight.177530. Pathogenic variants in SCN8A, which encodes the voltage-gated sodium (NaV) channel NaV1.6, associate with neurodevelopmental disorders, including developmental and epileptic encephalopathy. Previous approaches to determine SCN8A variant function may be confounded by use of a neonatally expressed, alternatively spliced isoform of NaV1.6 (NaV1.6N) and engineered mutations rendering the channel tetrodotoxin (TTX) resistant. We investigated the impact of SCN8A alternative splicing on variant function by comparing the functional attributes of 15 variants expressed in 2 developmentally regulated splice isoforms (NaV1.6N, NaV1.6A). We employed automated patch clamp recording to enhance throughput and developed a neuronal cell line (ND7/LoNav) with low levels of endogenous NaV current to obviate the need for TTX-resistance mutations. Expression of NaV1.6N or NaV1.6A in ND7/LoNav cells generated NaV currents with small, but significant, differences in voltage dependence of activation and inactivation. TTX-resistant versions of both isoforms exhibited significant functional differences compared with the corresponding WT channels. We demonstrated that many of the 15 disease-associated variants studied exhibited isoform-dependent functional effects and that many of the studied SCN8A variants exhibited functional properties that were not easily classified as either gain- or loss-of-function. Our work illustrates the value of considering molecular and cellular context when investigating SCN8A variants.
Collapse
Affiliation(s)
- Shruthi H Iyer
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine
| | - Kristina A Simeone
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine
| |
Collapse
|
6
|
Shabani K, Krupp J, Lemesre E, Lévy N, Tran H. Voltage-Gated Ion Channel Compensatory Effect in DEE: Implications for Future Therapies. Cells 2024; 13:1763. [PMID: 39513870 PMCID: PMC11544952 DOI: 10.3390/cells13211763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Developmental and Epileptic Encephalopathies (DEEs) represent a clinically and genetically heterogeneous group of rare and severe epilepsies. DEEs commonly begin early in infancy with frequent seizures of various types associated with intellectual disability and leading to a neurodevelopmental delay or regression. Disease-causing genomic variants have been identified in numerous genes and are implicated in over 100 types of DEEs. In this context, genes encoding voltage-gated ion channels (VGCs) play a significant role, and part of the large phenotypic variability observed in DEE patients carrying VGC mutations could be explained by the presence of genetic modifier alleles that can compensate for these mutations. This review will focus on the current knowledge of the compensatory effect of DEE-associated voltage-gated ion channels and their therapeutic implications in DEE. We will enter into detailed considerations regarding the sodium channels SCN1A, SCN2A, and SCN8A; the potassium channels KCNA1, KCNQ2, and KCNT1; and the calcium channels CACNA1A and CACNA1G.
Collapse
Affiliation(s)
- Khadijeh Shabani
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| | | | | | | | - Helene Tran
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| |
Collapse
|
7
|
Rubio C, Romo-Parra H, López-Landa A, Rubio-Osornio M. Classification of Current Experimental Models of Epilepsy. Brain Sci 2024; 14:1024. [PMID: 39452036 PMCID: PMC11506208 DOI: 10.3390/brainsci14101024] [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: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
INTRODUCTION This article provides an overview of several experimental models, including in vivo, genetics, chemical, knock-in, knock-out, electrical, in vitro, and optogenetics models, that have been employed to investigate epileptogenesis. The present review introduces a novel categorization of these models, taking into account the fact that the most recent classification that gained widespread acceptance was established by Fisher in 1989. A significant number of such models have become virtually outdated. OBJECTIVE This paper specifically examines the models that have contributed to the investigation of partial seizures, generalized seizures, and status epilepticus. DISCUSSION A description is provided of the primary features associated with the processes that produce and regulate the symptoms of various epileptogenesis models. Numerous experimental epilepsy models in animals have made substantial contributions to the investigation of particular brain regions that are capable of inducing seizures. Experimental models of epilepsy have also enabled the investigation of the therapeutic mechanisms of anti-epileptic medications. Typically, animals are selected for the development and study of experimental animal models of epilepsy based on the specific form of epilepsy being investigated. CONCLUSIONS Currently, it is established that specific animal species can undergo epileptic seizures that resemble those described in humans. Nevertheless, it is crucial to acknowledge that a comprehensive assessment of all forms of human epilepsy has not been feasible. However, these experimental models, both those derived from channelopathies and others, have provided a limited comprehension of the fundamental mechanisms of this disease.
Collapse
Affiliation(s)
- Carmen Rubio
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
| | - Héctor Romo-Parra
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
- Psychology Department, Universidad Iberoamericana, Mexico City 01219, Mexico
| | - Alejandro López-Landa
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
| | - Moisés Rubio-Osornio
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía, Av. Insurgentes Sur 3877, Mexico City 14269, Mexico
| |
Collapse
|
8
|
Breimann S, Kamp F, Steiner H, Frishman D. AAontology: An Ontology of Amino Acid Scales for Interpretable Machine Learning. J Mol Biol 2024; 436:168717. [PMID: 39053689 DOI: 10.1016/j.jmb.2024.168717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Amino acid scales are crucial for protein prediction tasks, many of them being curated in the AAindex database. Despite various clustering attempts to organize them and to better understand their relationships, these approaches lack the fine-grained classification necessary for satisfactory interpretability in many protein prediction problems. To address this issue, we developed AAontology-a two-level classification for 586 amino acid scales (mainly from AAindex) together with an in-depth analysis of their relations-using bag-of-word-based classification, clustering, and manual refinement over multiple iterations. AAontology organizes physicochemical scales into 8 categories and 67 subcategories, enhancing the interpretability of scale-based machine learning methods in protein bioinformatics. Thereby it enables researchers to gain a deeper biological insight. We anticipate that AAontology will be a building block to link amino acid properties with protein function and dysfunctions as well as aid informed decision-making in mutation analysis or protein drug design.
Collapse
Affiliation(s)
- Stephan Breimann
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany; Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Frits Kamp
- Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany
| | - Harald Steiner
- Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Dmitrij Frishman
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
9
|
Egido-Betancourt HX, Strowd III RE, Raab-Graham KF. Potential roles of voltage-gated ion channel disruption in Tuberous Sclerosis Complex. Front Mol Neurosci 2024; 17:1404884. [PMID: 39253727 PMCID: PMC11381416 DOI: 10.3389/fnmol.2024.1404884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 09/11/2024] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a lynchpin disorder, as it results in overactive mammalian target of rapamycin (mTOR) signaling, which has been implicated in a multitude of disease states. TSC is an autosomal dominant disease where 90% of affected individuals develop epilepsy. Epilepsy results from aberrant neuronal excitability that leads to recurring seizures. Under neurotypical conditions, the coordinated activity of voltage-gated ion channels keep neurons operating in an optimal range, thus providing network stability. Interestingly, loss or gain of function mutations in voltage-gated potassium, sodium, or calcium channels leads to altered excitability and seizures. To date, little is known about voltage-gated ion channel expression and function in TSC. However, data is beginning to emerge on how mTOR signaling regulates voltage-gated ion channel expression in neurons. Herein, we provide a comprehensive review of the literature describing common seizure types in patients with TSC, and suggest possible parallels between acquired epilepsies with known voltage-gated ion channel dysfunction. Furthermore, we discuss possible links toward mTOR regulation of voltage-gated ion channels expression and channel kinetics and the underlying epileptic manifestations in patients with TSC.
Collapse
Affiliation(s)
- Hailey X. Egido-Betancourt
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Roy E. Strowd III
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
10
|
Zhang W, Mi S, He X, Cui J, Zhi K, Zhang J. Advancements in Non-Addictive Analgesic Diterpenoid Alkaloid Lappaconitine: A Review. Int J Mol Sci 2024; 25:8255. [PMID: 39125825 PMCID: PMC11311510 DOI: 10.3390/ijms25158255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/13/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
The perennial herb Aconitum sinomontanum Nakai (Ranunculaceae) has been utilized as a traditional oriental medicine in China for numerous years. The principal pharmacological constituent of A. sinomontanum, lappaconitine (LA), exhibits analgesic, anti-inflammatory, anti-tumor, anti-arrhythmic, and anti-epileptic activities. Due to its potent efficacy and non-addictive nature, LA is widely utilized in the management of cancer pain and postoperative analgesia. This review encompasses the research advancements pertaining to LA including extraction methods, separation techniques, pharmacological properties, chemical modifications, and clinical applications. Additionally, it offers insights into the potential applications and current challenges associated with LA to facilitate future research endeavors.
Collapse
Affiliation(s)
- Wen Zhang
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
- Institute of New Rural Development, Northwest Normal University, Lanzhou 730070, China
| | - Shujuan Mi
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
| | - Xinxin He
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
| | - Jiajia Cui
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
- Institute of New Rural Development, Northwest Normal University, Lanzhou 730070, China
| | - Kangkang Zhi
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
- Institute of New Rural Development, Northwest Normal University, Lanzhou 730070, China
| | - Ji Zhang
- College of Life Science, Northwest Normal University, Lanzhou 730070, China; (S.M.); (X.H.); (J.C.); (K.Z.)
- Institute of New Rural Development, Northwest Normal University, Lanzhou 730070, China
| |
Collapse
|
11
|
Duma GM, Cuozzo S, Wilson L, Danieli A, Bonanni P, Pellegrino G. Excitation/Inhibition balance relates to cognitive function and gene expression in temporal lobe epilepsy: a high density EEG assessment with aperiodic exponent. Brain Commun 2024; 6:fcae231. [PMID: 39056027 PMCID: PMC11272395 DOI: 10.1093/braincomms/fcae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/22/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Patients with epilepsy are characterized by a dysregulation of excitation/inhibition balance (E/I). The assessment of E/I may inform clinicians during the diagnosis and therapy management, even though it is rarely performed. An accessible measure of the E/I of the brain represents a clinically relevant feature. Here, we exploited the exponent of the aperiodic component of the power spectrum of the electroencephalography (EEG) signal, as a non-invasive and cost-effective proxy of the E/I balance. We recorded resting-state activity with high-density EEG from 67 patients with temporal lobe epilepsy and 35 controls. We extracted the exponent of the aperiodic fit of the power spectrum from source-reconstructed EEG and tested differences between patients with epilepsy and controls. Spearman's correlation was performed between the exponent and clinical variables (age of onset, epilepsy duration and neuropsychology) and cortical expression of epilepsy-related genes derived from the Allen Human Brain Atlas. Patients with temporal lobe epilepsy showed a significantly larger exponent, corresponding to inhibition-directed E/I balance, in bilateral frontal and temporal regions. Lower E/I in the left entorhinal and bilateral dorsolateral prefrontal cortices corresponded to a lower performance of short-term verbal memory. Limited to patients with temporal lobe epilepsy, we detected a significant correlation between the exponent and the cortical expression of GABRA1, GRIN2A, GABRD, GABRG2, KCNA2 and PDYN genes. EEG aperiodic exponent maps the E/I balance non-invasively in patients with epilepsy and reveals a close relationship between altered E/I patterns, cognition and genetics.
Collapse
Affiliation(s)
- Gian Marco Duma
- Scientific Institute IRCCS E.Medea, Epilepsy and Clinical Neurophysiology Unit, 31015, Conegliano, Italy
| | - Simone Cuozzo
- Scientific Institute IRCCS E.Medea, Epilepsy and Clinical Neurophysiology Unit, 31015, Conegliano, Italy
| | - Luc Wilson
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alberto Danieli
- Scientific Institute IRCCS E.Medea, Epilepsy and Clinical Neurophysiology Unit, 31015, Conegliano, Italy
| | - Paolo Bonanni
- Scientific Institute IRCCS E.Medea, Epilepsy and Clinical Neurophysiology Unit, 31015, Conegliano, Italy
| | - Giovanni Pellegrino
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London N6A5C1, Canada
| |
Collapse
|
12
|
Shin HJ, Ko A, Kim SH, Lee JS, Kang HC. Unusual Voltage-Gated Sodium and Potassium Channelopathies Related to Epilepsy. J Clin Neurol 2024; 20:402-411. [PMID: 38951973 PMCID: PMC11220354 DOI: 10.3988/jcn.2023.0435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND AND PURPOSE There is extensive literature on monogenic epilepsies caused by mutations in familiar channelopathy genes such as SCN1A. However, information on other less-common channelopathy genes is scarce. This study aimed to explore the genetic and clinical characteristics of patients diagnosed with unusual voltage-gated sodium and potassium channelopathies related to epilepsy. METHODS This observational, retrospective study analyzed pediatric patients with epilepsy who carried pathogenic variants of unusual voltage-gated sodium and potassium channelopathy genes responsible for seizure-associated phenotypes. Targeted next-generation sequencing (NGS) panel tests were performed between November 2016 and June 2022 at Severance Children's Hospital, Seoul, South Korea. Clinical characteristics and the treatment responses to different types of antiseizure medications were further analyzed according to different types of gene mutation. RESULTS This study included 15 patients with the following unusual voltage-gated sodium and potassium channelopathy genes: SCN3A (n=1), SCN4A (n=1), KCNA1 (n=1), KCNA2 (n=4), KCNB1 (n=6), KCNC1 (n=1), and KCNMA1 (n=1). NGS-based genetic testing identified 13 missense mutations (87%), 1 splice-site variant (7%), and 1 copy-number variant (7%). Developmental and epileptic encephalopathy was diagnosed in nine (60%) patients. Seizure freedom was eventually achieved in eight (53%) patients, whereas seizures persisted in seven (47%) patients. CONCLUSIONS Our findings broaden the genotypic and phenotypic spectra of less-common voltage-gated sodium and potassium channelopathies associated with epilepsy.
Collapse
Affiliation(s)
- Hui Jin Shin
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ara Ko
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hee Kim
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Joon Soo Lee
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
13
|
Pant K, Sharma A, Menon SV, Ali H, Hassan Almalki W, Kaur M, Deorari M, Kazmi I, Mahajan S, Kalra H, Alzarea SI. Exploring ncRNAs in epilepsy: From oxidative stress regulation to therapy. Brain Res 2024; 1841:149089. [PMID: 38880410 DOI: 10.1016/j.brainres.2024.149089] [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: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Epilepsy is a prevalent neurological illness which is linked with high worldwide burdens. Oxidative stress (OS) is recognized to be among the contributors that trigger the advancement of epilepsy, affecting neuronal excitability and synaptic transmission. Various types of non-coding RNAs (ncRNAs) are known to serve vital functions in many disease mechanisms, including epilepsy. The current review sought to understand better the mechanisms through which these ncRNAs regulate epilepsy's OS-related pathways. We investigated the functions of microRNAs in controlling gene expression at the post-translatory stage and their involvement in OS and neuroinflammation. We also looked at the different regulatory roles of long ncRNAs, including molecular scaffolding, enhancer, and transcriptional activator, during OS. Circular RNAs and their capability to act as miRNA decoys and their consequential impact on epilepsy development were also explored. Our review aimed to improve the current understanding of novel therapies for epilepsy based on the role of ncRNAs in OS pathways. We also demonstrated the roles of ncRNAs in epilepsy treatment and diagnosis, explaining that these molecules play vital roles that could be used in therapy as biomarkers.
Collapse
Affiliation(s)
- Kumud Pant
- Graphic Era (Deemed to be University), Clement Town Dehradun, 248002, India; Graphic Era Hill University Clement Town Dehradun, 248002, India
| | - Aanchal Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali 140307, Punjab, India
| | - Soumya V Menon
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan.
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Mahamedha Deorari
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand- 831001, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Shriya Mahajan
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India
| | - Hitesh Kalra
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh 174103, India
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| |
Collapse
|
14
|
Córdoba NM, Lince-Rivera I, Gómez JLR, Rubboli G, De la Rosa SO. ATP1A2-related epileptic encephalopathy and movement disorder: Clinical features of three novel patients. Epileptic Disord 2024; 26:332-340. [PMID: 38512072 DOI: 10.1002/epd2.20220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
OBJECTIVE Variants in the ATP1A2 gene exhibit a wide clinical spectrum, ranging from familial hemiplegic migraine to childhood epilepsies and early infantile developmental epileptic encephalopathy (EIDEE) with movement disorders. This study aims to describe the epileptology of three unpublished cases and summarize epilepsy features of the other 17 published cases with ATP1A2 variants and EIDEE. METHODS Medical records of three novel patients with pathogenic ATP1A2 variants were retrospectively reviewed. Additionally, the PUBMED, EMBASE, and Cochrane databases were searched until December 2023 for articles on EIDEE with ATP1A2 variants, without language or publication year restrictions. RESULTS Three female patients, aged 6 months-10 years, were investigated. Epilepsy onset occurred between 5 days and 2 years, accompanied by severe developmental delay, intellectual disability, drug-resistant epilepsy, severe movement disorder, and recurrent status epilepticus. All individuals had pathogenic variants of the ATP1A2 gene (ATP1A2 c.720_721del (p.Ile240MetfsTer9), ATP1A2c.3022C > T (p.Arg1008Trp), ATP1A2 c.1096G > T (p.Gly366Cys), according to ACMG criteria. Memantine was p) rescribed to three patients, one with a reduction in ictal frequency, one with improvement in gait pattern, coordination, and attention span, and another one in alertness without significant side effects. SIGNIFICANCE This study reinforces the association between ATP1A2 variants and a severe phenotype. All patients had de novo variants, focal motor seizures with impaired awareness as the primary type of seizure; of the 11 EEGs recorded, 10 presented a slow background rhythm, 7 multifocal interictal epileptiform discharges (IED), predominantly temporal IEDs, followed by frontal IED, as well as ten ictal recordings, which showed ictal onset from the same regions mentioned above. Treatment with antiseizure medication was generally ineffective, but memantine showed moderate improvement. Prospective studies are needed to enlarge the phenotype and assess the efficacy of NMDA receptor antagonist therapies in reducing seizure frequency and improving quality of life.
Collapse
Affiliation(s)
| | | | | | - Guido Rubboli
- Danish Epilepsy Center, Member of ERN EpiCARE, Dianalund, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sebastián Ortiz De la Rosa
- Instituto Roosevelt, Bogotá, Colombia
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
- Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
15
|
Quinn S, Zhang N, Fenton TA, Brusel M, Muruganandam P, Peleg Y, Giladi M, Haitin Y, Lerche H, Bassan H, Liu Y, Ben-Shalom R, Rubinstein M. Complex biophysical changes and reduced neuronal firing in an SCN8A variant associated with developmental delay and epilepsy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167127. [PMID: 38519006 DOI: 10.1016/j.bbadis.2024.167127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024]
Abstract
Mutations in the SCN8A gene, encoding the voltage-gated sodium channel NaV1.6, are associated with a range of neurodevelopmental syndromes. The p.(Gly1625Arg) (G1625R) mutation was identified in a patient diagnosed with developmental epileptic encephalopathy (DEE). While most of the characterized DEE-associated SCN8A mutations were shown to cause a gain-of-channel function, we show that the G1625R variant, positioned within the S4 segment of domain IV, results in complex effects. Voltage-clamp analyses of NaV1.6G1625R demonstrated a mixture of gain- and loss-of-function properties, including reduced current amplitudes, increased time constant of fast voltage-dependent inactivation, a depolarizing shift in the voltage dependence of activation and inactivation, and increased channel availability with high-frequency repeated depolarization. Current-clamp analyses in transfected cultured neurons revealed that these biophysical properties caused a marked reduction in the number of action potentials when firing was driven by the transfected mutant NaV1.6. Accordingly, computational modeling of mature cortical neurons demonstrated a mild decrease in neuronal firing when mimicking the patients' heterozygous SCN8A expression. Structural modeling of NaV1.6G1625R suggested the formation of a cation-π interaction between R1625 and F1588 within domain IV. Double-mutant cycle analysis revealed that this interaction affects the voltage dependence of inactivation in NaV1.6G1625R. Together, our studies demonstrate that the G1625R variant leads to a complex combination of gain and loss of function biophysical changes that result in an overall mild reduction in neuronal firing, related to the perturbed interaction network within the voltage sensor domain, necessitating personalized multi-tiered analysis for SCN8A mutations for optimal treatment selection.
Collapse
Affiliation(s)
- Shir Quinn
- Goldschleger Eye Research Institute, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nan Zhang
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Timothy A Fenton
- Neurology Department, MIND Institute, University of California, Davis, Sacramento, CA, United States
| | - Marina Brusel
- Goldschleger Eye Research Institute, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Preethi Muruganandam
- Neurology Department, MIND Institute, University of California, Davis, Sacramento, CA, United States
| | - Yoav Peleg
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Giladi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yoni Haitin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Haim Bassan
- Pediatric Neurology and Development Center, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.
| | - Roy Ben-Shalom
- Neurology Department, MIND Institute, University of California, Davis, Sacramento, CA, United States.
| | - Moran Rubinstein
- Goldschleger Eye Research Institute, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Elkhateeb N, Issa MY, Elbendary HM, Elnaggar W, Ramadan A, Rafat K, Kamel M, Abdel-Ghafar SF, Amer F, Hassaan HM, Trunzo R, Pereira C, Abdel-Hamid MS, D'Arco F, Bauer P, Bertoli-Avella AM, Girgis M, Gleeson JG, Zaki MS, Selim L. The clinical and genetic landscape of developmental and epileptic encephalopathies in Egyptian children. Clin Genet 2024; 105:510-522. [PMID: 38221827 DOI: 10.1111/cge.14481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/16/2024]
Abstract
Developmental and epileptic encephalopathies (DEEs) are a heterogeneous group of epilepsies characterized by early-onset, refractory seizures associated with developmental regression or impairment, with a heterogeneous genetic landscape including genes implicated in various pathways and mechanisms. We retrospectively studied the clinical and genetic data of patients with genetic DEE who presented at two tertiary centers in Egypt over a 10-year period. Exome sequencing was used for genetic testing. We report 74 patients from 63 unrelated Egyptian families, with a high rate of consanguinity (58%). The most common seizure type was generalized tonic-clonic (58%) and multiple seizure types were common (55%). The most common epilepsy syndrome was early infantile DEE (50%). All patients showed variable degrees of developmental impairment. Microcephaly, hypotonia, ophthalmological involvement and neuroimaging abnormalities were common. Eighteen novel variants were identified and the phenotypes of five DEE genes were expanded with novel phenotype-genotype associations. Obtaining a genetic diagnosis had implications on epilepsy management in 17 patients with variants in 12 genes. In this study, we expand the phenotype and genotype spectrum of DEE in a large single ethnic cohort of patients. Reaching a genetic diagnosis guided precision management of epilepsy in a significant proportion of patients.
Collapse
Affiliation(s)
- Nour Elkhateeb
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Mahmoud Y Issa
- Department of Clinical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Hasnaa M Elbendary
- Department of Clinical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Walaa Elnaggar
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Areef Ramadan
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Karima Rafat
- Department of Clinical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Mona Kamel
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Sherif F Abdel-Ghafar
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Fawzia Amer
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Hebatallah M Hassaan
- Department of Pediatrics, Clinical Genetics Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Mohamed S Abdel-Hamid
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Felice D'Arco
- Radiology Department, Great Ormond Street Hospital for Children, London, UK
| | | | | | - Marian Girgis
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Joseph G Gleeson
- Department of Neurosciences, University of California, San Diego, La Jolla, USA
- Rady Children's Hospital, Rady Children's Institute for Genomic Medicine, San Diego, La Jolla, USA
| | - Maha S Zaki
- Department of Clinical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Laila Selim
- Department of Pediatrics, Pediatric Neurology and Metabolic Medicine Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
17
|
Zhao K, Li Y, Lai H, Niu R, Li H, He S, Su Z, Gui Y, Ren L, Yang X, Zhou L. Alterations in HCN1 expression and distribution during epileptogenesis in rats. Epilepsy Res 2024; 202:107355. [PMID: 38555654 DOI: 10.1016/j.eplepsyres.2024.107355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The hyperpolarization-activated cyclic nucleotide-gated cation channel (HCN1) is predominantly located in key regions associated with epilepsy, such as the neocortex and hippocampus. Under normal physiological conditions, HCN1 plays a crucial role in the excitatory and inhibitory regulation of neuronal networks. In temporal lobe epilepsy, the expression of HCN1 is decreased in the hippocampi of both animal models and patients. However, whether HCN1 expression changes during epileptogenesis preceding spontaneous seizures remains unclear. OBJECTIVE The aim of this study was to determine whether the expression of HCN1 is altered during the epileptic prodromal phase, thereby providing evidence for its role in epileptogenesis. METHODS We utilized a cobalt wire-induced rat epilepsy model to observe changes in HCN1 during epileptogenesis and epilepsy. Additionally, we also compared HCN1 alterations in epileptogenic tissues between cobalt wire- and pilocarpine-induced epilepsy rat models. Long-term video EEG recordings were used to confirm seizures development. Transcriptional changes, translation, and distribution of HCN1 were assessed using high-throughput transcriptome sequencing, total protein extraction, membrane and cytoplasmic protein fractionation, western blotting, immunohistochemistry, and immunofluorescence techniques. RESULTS In the cobalt wire-induced rat epilepsy model during the epileptogenesis phase, total HCN1 mRNA and protein levels were downregulated. Specifically, the membrane expression of HCN1 was decreased, whereas cytoplasmic HCN1 expression showed no significant change. The distribution of HCN1 in the distal dendrites of neurons decreased. During the epilepsy period, similar HCN1 alterations were observed in the neocortex of rats with cobalt wire-induced epilepsy and hippocampus of rats with lithium pilocarpine-induced epilepsy, including downregulation of mRNA levels, decreased total protein expression, decreased membrane expression, and decreased distal dendrite expression. CONCLUSIONS Alterations in HCN1 expression and distribution are involved in epileptogenesis beyond their association with seizure occurrence. Similarities in HCN1 alterations observed in epileptogenesis-related tissues from different models suggest a shared pathophysiological pathway in epileptogenesis involving HCN1 dysregulation. Therefore, the upregulation of HCN1 expression in neurons, maintenance of the HCN1 membrane, and distal dendrite distribution in neurons may represent promising disease-modifying strategies in epilepsy.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Guangzhou National Laboratory, Guangzhou, China; Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yinchao Li
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | | | - Ruili Niu
- Guangzhou National Laboratory, Guangzhou, China
| | - Huifeng Li
- Guangzhou National Laboratory, Guangzhou, China
| | - Shipei He
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhengwei Su
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yue Gui
- Guangzhou National Laboratory, Guangzhou, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
| | | | - Liemin Zhou
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
18
|
Dell'Isola GB, Verrotti A, Sciaccaluga M, Dini G, Ferrara P, Parnetti L, Costa C. Cannabidiol: metabolism and clinical efficacy in epileptic patients. Expert Opin Drug Metab Toxicol 2024; 20:119-131. [PMID: 38465404 DOI: 10.1080/17425255.2024.2329733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION The landscape of epilepsy treatment has undergone a significant transformation with the emergence of cannabidiol as a potential therapeutic agent. Epidiolex, a pharmaceutical formulation of highly purified CBD, garnered significant attention not just for its therapeutic potential but also for being the first cannabis-derived medication to obtain approval from regulatory bodies. AREA COVERED In this narrative review the authors explore the intricate landscape of CBD as an antiseizure medication, deepening into its pharmacological mechanisms and clinical trials involving various epileptic encephalopathies. This exploration serves as a comprehensive guide, shedding light on a compound that holds promise for individuals contending with the significant challenges of drug-resistant epilepsy. EXPERT OPINION Rigorous studies highlight cannabidiol's efficacy, safety profile, and potential cognitive benefits, warranting further exploration for its approval in various drug-resistant epilepsy forms. As a promising therapeutic option, cannabidiol not only demonstrates efficacy in seizure control but also holds the potential for broader enhancements in the quality of life, especially for patients with epileptic encephalopathies.
Collapse
Affiliation(s)
| | | | - Miriam Sciaccaluga
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- "Mauro Baschirotto" Institute for Rare Diseases - BIRD Foundation Onlus, Longare, Vicenza, Italy
| | - Gianluca Dini
- Department of Pediatrics, University of Perugia, Perugia, Italy
| | - Pietro Ferrara
- Unit of Pediatrics, Campus Bio-Medico University, Rome, Italy
| | - Lucilla Parnetti
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Costa
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
Chen Y, Mateski J, Gerace L, Wheeler J, Burl J, Prakash B, Svedin C, Amrick R, Adams BD. Non-coding RNAs and neuroinflammation: implications for neurological disorders. Exp Biol Med (Maywood) 2024; 249:10120. [PMID: 38463392 PMCID: PMC10911137 DOI: 10.3389/ebm.2024.10120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 03/12/2024] Open
Abstract
Neuroinflammation is considered a balanced inflammatory response important in the intrinsic repair process after injury or infection. Under chronic states of disease, injury, or infection, persistent neuroinflammation results in a heightened presence of cytokines, chemokines, and reactive oxygen species that result in tissue damage. In the CNS, the surrounding microglia normally contain macrophages and other innate immune cells that perform active immune surveillance. The resulting cytokines produced by these macrophages affect the growth, development, and responsiveness of the microglia present in both white and gray matter regions of the CNS. Controlling the levels of these cytokines ultimately improves neurocognitive function and results in the repair of lesions associated with neurologic disease. MicroRNAs (miRNAs) are master regulators of the genome and subsequently control the activity of inflammatory responses crucial in sustaining a robust and acute immunological response towards an acute infection while dampening pathways that result in heightened levels of cytokines and chemokines associated with chronic neuroinflammation. Numerous reports have directly implicated miRNAs in controlling the abundance and activity of interleukins, TGF-B, NF-kB, and toll-like receptor-signaling intrinsically linked with the development of neurological disorders such as Parkinson's, ALS, epilepsy, Alzheimer's, and neuromuscular degeneration. This review is focused on discussing the role miRNAs play in regulating or initiating these chronic neurological states, many of which maintain the level and/or activity of neuron-specific secondary messengers. Dysregulated miRNAs present in the microglia, astrocytes, oligodendrocytes, and epididymal cells, contribute to an overall glial-specific inflammatory niche that impacts the activity of neuronal conductivity, signaling action potentials, neurotransmitter robustness, neuron-neuron specific communication, and neuron-muscular connections. Understanding which miRNAs regulate microglial activation is a crucial step forward in developing non-coding RNA-based therapeutics to treat and potentially correct the behavioral and cognitive deficits typically found in patients suffering from chronic neuroinflammation.
Collapse
Affiliation(s)
- Yvonne Chen
- Department of Biology, Brandeis University, Waltham, MA, United States
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
| | - Julia Mateski
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Biological Sciences, Gustavus Adolphus College, St. Peter, MN, United States
| | - Linda Gerace
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Missouri State University, Springfield, MO, United States
| | - Jonathan Wheeler
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Electrical and Computer Engineering Tech, New York Institute of Tech, Old Westbury, NY, United States
| | - Jan Burl
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Southern New Hampshire University, Manchester, NH, United States
| | - Bhavna Prakash
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Medicine, Tufts Medical Center, Medford, MA, United States
| | - Cherie Svedin
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Biology, Utah Tech University, St. George, UT, United States
| | - Rebecca Amrick
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Villanova University, Villanova, PA, United States
| | - Brian D Adams
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
| |
Collapse
|
20
|
Snyder HE, Jain P, RamachandranNair R, Jones KC, Whitney R. Genetic Advancements in Infantile Epileptic Spasms Syndrome and Opportunities for Precision Medicine. Genes (Basel) 2024; 15:266. [PMID: 38540325 PMCID: PMC10970414 DOI: 10.3390/genes15030266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 06/15/2024] Open
Abstract
Infantile epileptic spasms syndrome (IESS) is a devastating developmental epileptic encephalopathy (DEE) consisting of epileptic spasms, as well as one or both of developmental regression or stagnation and hypsarrhythmia on EEG. A myriad of aetiologies are associated with the development of IESS; broadly, 60% of cases are thought to be structural, metabolic or infectious in nature, with the remainder genetic or of unknown cause. Epilepsy genetics is a growing field, and over 28 copy number variants and 70 single gene pathogenic variants related to IESS have been discovered to date. While not exhaustive, some of the most commonly reported genetic aetiologies include trisomy 21 and pathogenic variants in genes such as TSC1, TSC2, CDKL5, ARX, KCNQ2, STXBP1 and SCN2A. Understanding the genetic mechanisms of IESS may provide the opportunity to better discern IESS pathophysiology and improve treatments for this condition. This narrative review presents an overview of our current understanding of IESS genetics, with an emphasis on animal models of IESS pathogenesis, the spectrum of genetic aetiologies of IESS (i.e., chromosomal disorders, single-gene disorders, trinucleotide repeat disorders and mitochondrial disorders), as well as available genetic testing methods and their respective diagnostic yields. Future opportunities as they relate to precision medicine and epilepsy genetics in the treatment of IESS are also explored.
Collapse
Affiliation(s)
- Hannah E. Snyder
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Puneet Jain
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Rajesh RamachandranNair
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Kevin C. Jones
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| |
Collapse
|
21
|
Kwan R, Das P, Gerrebos N, Li J, Wang XY, DeBoer G, Emnacen-Pankhurst V, Lin S, Feng R, Goodchild S, Sojo LE. Development and application of a multiple reaction monitoring method for the simultaneous quantification of sodium channels Na v 1.1, Na v 1.2, and Na v 1.6 in solubilized membrane proteins from stable HEK293 cell lines, rodents, and human brain tissues. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9672. [PMID: 38211346 DOI: 10.1002/rcm.9672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/13/2023] [Accepted: 10/22/2023] [Indexed: 01/13/2024]
Abstract
RATIONALE Nav 1.1, 1.2, and 1.6 are transmembrane proteins acting as voltage-gated sodium channels implicated in various forms of epilepsy. There is a need for knowing their actual concentration in target tissues during drug development. METHODS Unique peptides for Nav 1.1, Nav 1.2, and Nav 1.6 were selected as quantotropic peptides for each protein and used for their quantification in membranes from stably transfected HEK293 cells and rodent and human brain samples using ultra-high-performance liquid chromatography-electrospray ionization tandem mass spectrometry. RESULTS Nav 1.1, 1.2, and 1.6 protein expressions in three stably individually transfected HEK293 cell lines were found to be 2.1 ± 0.2, 6.4 ± 1.2, and 4.0 ± 0.6 fmol/μg membrane protein, respectively. In brains, Nav 1.2 showed the highest expression, with approximately three times higher (P < 0.003) in rodents than in humans at 3.05 ± 0.57, with 3.35 ± 0.56 in mouse and rat brains and 1.09 ± 0.27 fmol/μg in human brain. Both Nav 1.1 and 1.6 expressions were much lower in the brains, with approximately 40% less expression in human Nav 1.1 than rodent Nav 1.1 at 0.49 ± 0.1 (mouse), 0.43 ± 0.3 (rat), and 0.28 ± 0.04 (humans); whereas Nav 1.6 had approximately 60% less expression in humans than rodents at 0.27 ± 0.09 (mouse), 0.26 ± 0.06 (rat), and 0.11 ± 0.02 (humans) fmol/μg membrane proteins. CONCLUSIONS Multiple reaction monitoring was used to quantify sodium channels Nav 1.1, 1.2, and 1.6 expressed in stably transfected HEK293 cells and brain tissues from mice, rats, and humans. We found significant differences in the expression of these channels in mouse, rat, and human brains. Nav expression ranking among the three species was Nav 1.2 ≫ Nav 1.1 > Nav 1.6, with the human brain expressing much lower concentrations overall compared to rodent brain.
Collapse
Affiliation(s)
- Rainbow Kwan
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Prerna Das
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Neelan Gerrebos
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Jenny Li
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Xin Yin Wang
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Gina DeBoer
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | | | - Sophia Lin
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Raymond Feng
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Sam Goodchild
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| | - Luis E Sojo
- Xenon Pharmaceuticals Inc., Burnaby, British Columbia, Canada
| |
Collapse
|
22
|
Hung TY, Wu SN, Huang CW. Safinamide, an inhibitor of monoamine oxidase, modulates the magnitude, gating, and hysteresis of sodium ion current. BMC Pharmacol Toxicol 2024; 25:17. [PMID: 38331833 PMCID: PMC10851555 DOI: 10.1186/s40360-024-00739-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Safinamide (SAF), an α-aminoamide derivative and a selective, reversible monoamine oxidase (MAO)-B inhibitor, has both dopaminergic and nondopaminergic (glutamatergic) properties. Several studies have explored the potential of SAF against various neurological disorders; however, to what extent SAF modulates the magnitude, gating, and voltage-dependent hysteresis [Hys(V)] of ionic currents remains unknown. METHODS With the aid of patch-clamp technology, we investigated the effects of SAF on voltage-gated sodium ion (NaV) channels in pituitary GH3 cells. RESULTS SAF concentration-dependently stimulated the transient (peak) and late (sustained) components of voltage-gated sodium ion current (INa) in pituitary GH3 cells. The conductance-voltage relationship of transient INa [INa(T)] was shifted to more negative potentials with the SAF presence; however, the steady-state inactivation curve of INa(T) was shifted in a rightward direction in its existence. SAF increased the decaying time constant of INa(T) induced by a train of depolarizing stimuli. Notably, subsequent addition of ranolazine or mirogabalin reversed the SAF-induced increase in the decaying time constant. SAF also increased the magnitude of window INa induced by an ascending ramp voltage Vramp. Furthermore, SAF enhanced the Hys(V) behavior of persistent INa induced by an upright isosceles-triangular Vramp. Single-channel cell-attached recordings indicated SAF effectively increased the open-state probability of NaV channels. Molecular docking revealed SAF interacts with both MAO and NaV channels. CONCLUSION SAF may interact directly with NaV channels in pituitary neuroendocrine cells, modulating membrane excitability.
Collapse
Affiliation(s)
- Te-Yu Hung
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan.
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan.
- School of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
23
|
Sakaibara M, Yamamoto H, Murota H, Monma N, Sato S, Hirano-Iwata A. Enhanced responses to inflammatory cytokine interleukin-6 in micropatterned networks of cultured cortical neurons. Biochem Biophys Res Commun 2024; 695:149379. [PMID: 38159413 DOI: 10.1016/j.bbrc.2023.149379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
Cortical neurons in dissociated cultures are an indispensable model system for pharmacological research that provides insights into chemical responses in well-defined environments. However, cortical neurons plated on homogeneous substrates develop an unstructured network that exhibits excessively synchronized activity, which occasionally masks the consequences induced by external substances. Here, we show that hyperactivity and excessive synchrony in cultured cortical networks can be effectively suppressed by growing neurons in microfluidic devices. These devices feature a hierarchically modular design that resembles the in vivo network. We focused on interleukin-6, a pro-inflammatory cytokine, and assessed its acute and chronic effects. Fluorescence calcium imaging of spontaneous neural activity for up to 20 days of culture showed detectable modulation of collective activity events and neural correlation in micropatterned neurons, which was not apparent in neurons cultured on homogeneous substrates. Our results indicate that engineered neuronal networks provide a unique platform for detecting and understanding the fundamental effects of biochemical compounds on neuronal networks.
Collapse
Affiliation(s)
- Mamoru Sakaibara
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Hideaki Yamamoto
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan.
| | - Hakuba Murota
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Nobuaki Monma
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Shigeo Sato
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Ayumi Hirano-Iwata
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan; Advanced Institute for Materials Research, Tohoku University, Sendai, Japan; Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
24
|
Tang T, Li X, Yu E, Li M, Pan X. Identification of common core ion channel genes in epilepsy and Alzheimer's disease. Ir J Med Sci 2024; 193:417-424. [PMID: 37477849 DOI: 10.1007/s11845-023-03447-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Although available literature indicates that the incidence of dementia in the epilepsy population and the risk of seizures in the Alzheimer's disease (AD) population are high, the specific genetic risk factors and the interaction mechanism are unclear, rendering rational genetic interpretation rather challenging. AIMS Our work aims to identify the common core ion channel genes in epilepsy and AD. METHODS In this study, we first integrated gene expression omnibus datasets (GSE48350 and GSE6834) on AD and epilepsy to identify differentially expressed genes (DEGs), performing Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs. The related protein-protein interaction (PPI) network was constructed for DEGs, and the hub gene was evaluated. RESULTS A total of 2800 and 35 genes were identified in GSE48350 and GSE6834, and 12 DEGs were significantly differentially expressed between the datasets. KEGG pathway analysis showed that DEGs were primarily enriched in glutamatergic synapse and dopaminergic synapse pathways. SCN2A, GRIA1, and KCNJ9 were the hub genes with high connectivity. CONCLUSIONS The findings suggest that the three genes, SCN2A, GRIA1, and KCNJ9, may serve as potential targets for treating AD comorbid with epilepsy.
Collapse
Affiliation(s)
- Ting Tang
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Quanzhou, Fujian, 362000, People's Republic of China
| | - Xiang Li
- Department of Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, People's Republic of China
| | - Erhan Yu
- Department of Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, People's Republic of China
| | - Man Li
- Department of Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, People's Republic of China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, People's Republic of China.
| |
Collapse
|
25
|
Jiang L, Bi S, Lin L, He F, Deng F. Phenotypic and genetic characteristics of 24 cases of early infantile epileptic encephalopathy in East China, including a rare case of biallelic UGDH mutations. Mol Genet Genomic Med 2023; 11:e2269. [PMID: 37593999 PMCID: PMC10724516 DOI: 10.1002/mgg3.2269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/13/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Early infantile epileptic encephalopathy (EIEE) is a group of highly heterogeneous diseases, both phenotypically and genetically. Usually, it starts early on and manifests as intractable epilepsy, abnormal electroencephalogram, and growth retardation/intellectual impairment. With the advent of next-generation sequencing (NGS), its genetic etiology has attracted increasing clinical attention. This study aimed to investigate the genetic characteristics and clinical phenotypes of patients with EIEE from a central hospital in Eastern China. METHODS This study retrospectively included the gene variants from 24 EIEE-positive patients admitted between January 2021 and January 2022 to a hospital in Anhui Province, China. The genetic diagnosis was performed in all cases by trio-based whole-exome sequencing (WES). Additionally, Video electroencephalogram (VEEG) and neuroimaging examinations were performed. RESULTS A total of 24 children were included. The average age at the first seizure was approximately 5 months. About 42% of children had developmental retardation of varying degrees, 43% had brain structural abnormalities, and 64% had VEEG abnormalities. In addition, other phenotypes, including endocrine metabolism and cardiac structural abnormalities, have been independently reported. In total, fifteen pathogenic gene variants were identified in 24 patients. The main pathogenic genes identified were SCN1A (25%, 6/24), KCNQ2 (8.3%, 2/24), and TBC1D24 (8.3%, 2/24). We also found an extremely rare case of EIEE84 type caused by biallelic UGDH gene variants, predicting that this variant might affect the stability of the protein structure. CONCLUSIONS SCN1A pathogenic variants are the main factor leading to EIEE, similar to previously published cohort reports. NGS is useful for accurate clinical diagnoses and precise treatment choices. We also reported a rare case of EIEE84 caused by variants in the UGDH gene in a Chinese patient. This study further enriches the known spectrum of pathogenic EIEE genes.
Collapse
Affiliation(s)
- Liangliang Jiang
- Department of NeurologyChildren's Hospital of Anhui Medical University (Anhui Provincial Children's Hospital)HefeiChina
| | - Shaohua Bi
- Department of NeonatologyChildren's Hospital of Anhui Medical University (Anhui Provincial Children's Hospital)HefeiChina
| | - Li Lin
- Department of NeurologyChildren's Hospital of Anhui Medical University (Anhui Provincial Children's Hospital)HefeiChina
| | - Fan He
- Department of NeurologyChildren's Hospital of Anhui Medical University (Anhui Provincial Children's Hospital)HefeiChina
| | - Fang Deng
- Department of NephrologyChildren's Hospital of Anhui Medical University (Anhui Provincial Children's Hospital)HefeiChina
| |
Collapse
|
26
|
van Hugte EJH, Lewerissa EI, Wu KM, Scheefhals N, Parodi G, van Voorst TW, Puvogel S, Kogo N, Keller JM, Frega M, Schubert D, Schelhaas HJ, Verhoeven J, Majoie M, van Bokhoven H, Nadif Kasri N. SCN1A-deficient excitatory neuronal networks display mutation-specific phenotypes. Brain 2023; 146:5153-5167. [PMID: 37467479 PMCID: PMC10689919 DOI: 10.1093/brain/awad245] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023] Open
Abstract
Dravet syndrome is a severe epileptic encephalopathy, characterized by (febrile) seizures, behavioural problems and developmental delay. Eighty per cent of patients with Dravet syndrome have a mutation in SCN1A, encoding Nav1.1. Milder clinical phenotypes, such as GEFS+ (generalized epilepsy with febrile seizures plus), can also arise from SCN1A mutations. Predicting the clinical phenotypic outcome based on the type of mutation remains challenging, even when the same mutation is inherited within one family. This clinical and genetic heterogeneity adds to the difficulties of predicting disease progression and tailoring the prescription of anti-seizure medication. Understanding the neuropathology of different SCN1A mutations may help to predict the expected clinical phenotypes and inform the selection of best-fit treatments. Initially, the loss of Na+-current in inhibitory neurons was recognized specifically to result in disinhibition and consequently seizure generation. However, the extent to which excitatory neurons contribute to the pathophysiology is currently debated and might depend on the patient clinical phenotype or the specific SCN1A mutation. To examine the genotype-phenotype correlations of SCN1A mutations in relation to excitatory neurons, we investigated a panel of patient-derived excitatory neuronal networks differentiated on multi-electrode arrays. We included patients with different clinical phenotypes, harbouring various SCN1A mutations, along with a family in which the same mutation led to febrile seizures, GEFS+ or Dravet syndrome. We hitherto describe a previously unidentified functional excitatory neuronal network phenotype in the context of epilepsy, which corresponds to seizurogenic network prediction patterns elicited by proconvulsive compounds. We found that excitatory neuronal networks were affected differently, depending on the type of SCN1A mutation, but did not segregate according to clinical severity. Specifically, loss-of-function mutations could be distinguished from missense mutations, and mutations in the pore domain could be distinguished from mutations in the voltage sensing domain. Furthermore, all patients showed aggravated neuronal network responses at febrile temperatures compared with controls. Finally, retrospective drug screening revealed that anti-seizure medication affected GEFS+ patient- but not Dravet patient-derived neuronal networks in a patient-specific and clinically relevant manner. In conclusion, our results indicate a mutation-specific excitatory neuronal network phenotype, which recapitulates the foremost clinically relevant features, providing future opportunities for precision therapies.
Collapse
Affiliation(s)
- Eline J H van Hugte
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
- Department of Epileptology, ACE Kempenhaeghe, 5591 VE Heeze, The Netherlands
| | - Elly I Lewerissa
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Ka Man Wu
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Nicky Scheefhals
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Giulia Parodi
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, 16145 GE Genova, Italy
| | - Torben W van Voorst
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Sofia Puvogel
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Naoki Kogo
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Jason M Keller
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Monica Frega
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Helenius J Schelhaas
- Department of Neurology, Stichting Epilepsie Instellingen Nederland (SEIN), 2103 SW Heemstede, The Netherlands
| | - Judith Verhoeven
- Department of Epileptology, ACE Kempenhaeghe, 5591 VE Heeze, The Netherlands
| | - Marian Majoie
- Department of Epileptology, ACE Kempenhaeghe, 5591 VE Heeze, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
27
|
Llanos MA, Enrique N, Esteban-López V, Scioli-Montoto S, Sánchez-Benito D, Ruiz ME, Milesi V, López DE, Talevi A, Martín P, Gavernet L. A Combined Ligand- and Structure-Based Virtual Screening To Identify Novel NaV1.2 Blockers: In Vitro Patch Clamp Validation and In Vivo Anticonvulsant Activity. J Chem Inf Model 2023; 63:7083-7096. [PMID: 37917937 DOI: 10.1021/acs.jcim.3c00645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Epilepsy is a neurological disorder characterized by recurrent seizures that arise from abnormal electrical activity in the brain. Voltage-gated sodium channels (NaVs), responsible for the initiation and propagation of action potentials in neurons, play a critical role in the pathogenesis of epilepsy. This study sought to discover potential anticonvulsant compounds that interact with NaVs, specifically, the brain subtype hNaV1.2. A ligand-based QSAR model and a docking model were constructed, validated, and applied in a parallel virtual screening over the DrugBank database. Montelukast, Novobiocin, and Cinnarizine were selected for in vitro testing, using the patch-clamp technique, and all of them proved to inhibit hNaV1.2 channels heterologously expressed in HEK293 cells. Two hits were evaluated in the GASH/Sal model of audiogenic seizures and demonstrated promising activity, reducing the severity of sound-induced seizures at the doses tested. The combination of ligand- and structure-based models presents a valuable approach for identifying potential NaV inhibitors. These findings may provide a basis for further research into the development of new antiseizure drugs for the treatment of epilepsy.
Collapse
Affiliation(s)
- Manuel A Llanos
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Nicolás Enrique
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Vega Esteban-López
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Sebastian Scioli-Montoto
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - David Sánchez-Benito
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - María E Ruiz
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Veronica Milesi
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Dolores E López
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Alan Talevi
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Pedro Martín
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Luciana Gavernet
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| |
Collapse
|
28
|
Dongol Y, Wilson DT, Daly NL, Cardoso FC, Lewis RJ. Structure-function and rational design of a spider toxin Ssp1a at human voltage-gated sodium channel subtypes. Front Pharmacol 2023; 14:1277143. [PMID: 38034993 PMCID: PMC10682951 DOI: 10.3389/fphar.2023.1277143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
The structure-function and optimization studies of NaV-inhibiting spider toxins have focused on developing selective inhibitors for peripheral pain-sensing NaV1.7. With several NaV subtypes emerging as potential therapeutic targets, structure-function analysis of NaV-inhibiting spider toxins at such subtypes is warranted. Using the recently discovered spider toxin Ssp1a, this study extends the structure-function relationships of NaV-inhibiting spider toxins beyond NaV1.7 to include the epilepsy target NaV1.2 and the pain target NaV1.3. Based on these results and docking studies, we designed analogues for improved potency and/or subtype-selectivity, with S7R-E18K-rSsp1a and N14D-P27R-rSsp1a identified as promising leads. S7R-E18K-rSsp1a increased the rSsp1a potency at these three NaV subtypes, especially at NaV1.3 (∼10-fold), while N14D-P27R-rSsp1a enhanced NaV1.2/1.7 selectivity over NaV1.3. This study highlights the challenge of developing subtype-selective spider toxin inhibitors across multiple NaV subtypes that might offer a more effective therapeutic approach. The findings of this study provide a basis for further rational design of Ssp1a and related NaSpTx1 homologs targeting NaV1.2, NaV1.3 and/or NaV1.7 as research tools and therapeutic leads.
Collapse
Affiliation(s)
- Yashad Dongol
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David T. Wilson
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L. Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Fernanda C. Cardoso
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Richard J. Lewis
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Xia Z, He D, Wu Y, Kwok HF, Cao Z. Scorpion venom peptides: Molecular diversity, structural characteristics, and therapeutic use from channelopathies to viral infections and cancers. Pharmacol Res 2023; 197:106978. [PMID: 37923027 DOI: 10.1016/j.phrs.2023.106978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Animal venom is an important evolutionary innovation in nature. As one of the most representative animal venoms, scorpion venom contains an extremely diverse set of bioactive peptides. Scorpion venom peptides not only are 'poisons' that immobilize, paralyze, kill, or dissolve preys but also become important candidates for drug development and design. Here, the review focuses on the molecular diversity of scorpion venom peptides, their typical structural characteristics, and their multiple therapeutic or pharmaceutical applications in channelopathies, viral infections and cancers. Especially, the group of scorpion toxin TRPTx targeting transient receptor potential (TRP) channels is systematically summarized and worthy of attention because TRP channels play a crucial role in the regulation of homeostasis and the occurrence of diseases in human. We also further establish the potential relationship between the molecular characteristics and functional applications of scorpion venom peptides to provide a research basis for modern drug development and clinical utilization of scorpion venom resources.
Collapse
Affiliation(s)
- Zhiqiang Xia
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, China
| | - Dangui He
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macao.
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Bio-drug Research Center, Wuhan University, Wuhan, China.
| |
Collapse
|
30
|
Li MR, Luo XJ, Peng J. Role of sonic hedgehog signaling pathway in the regulation of ion channels: focus on its association with cardio-cerebrovascular diseases. J Physiol Biochem 2023; 79:719-730. [PMID: 37676576 DOI: 10.1007/s13105-023-00982-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Sonic hedgehog (SHH) signaling is vital for cell differentiation and proliferation during embryonic development, yet its role in cardiac, cerebral, and vascular pathophysiology is under debate. Recent studies have demonstrated that several compounds of SHH signaling regulate ion channels, which in turn affect the behavior of target cells. Some of these ion channels are involved in the cardio-cerebrovascular system. Here, we first reviewed the SHH signaling cascades, then its interaction with ion channels, and their impact on cardio-cerebrovascular diseases. Considering the complex cross talk of SHH signaling with other pathways that also affect ion channels and their potential impact on the cardio-cerebrovascular system, we highlight the necessity of thoroughly studying the effect of SHH signaling on ion homeostasis, which could serve as a novel mechanism for cardio-cerebrovascular diseases. Activation of SHH signaling influence ion channels activity, which in turn influence ion homeostasis, membrane potential, and electrophysiology, could serve as a novel strategy for cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
31
|
Asada S, Morita H, Mizuno T, Masuda T, Ueoka A, Miyamoto M, Kawada S, Nakagawa K, Nishii N. Syncope and loss of consciousness after implantation of a cardioverter-defibrillator in patients with Brugada syndrome: Prevalence and characteristics in long-term follow-up. Heart Rhythm O2 2023; 4:641-649. [PMID: 37936673 PMCID: PMC10626187 DOI: 10.1016/j.hroo.2023.09.007] [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] [Indexed: 11/09/2023] Open
Abstract
Background Syncope is a significant prognostic factor in patients with Brugada syndrome (BrS). However, the risk of ventricular arrhythmia in patients with nonarrhythmic loss of consciousness (LOC) is similar to that in asymptomatic patients. LOC events after implantable cardioverter-defibrillator (ICD) implantation may provide insights into underlying causes of the initial LOC episode. Objective The purpose of this study was to examine LOC characteristics following ICD implantation. Methods We retrospectively analyzed 112 patients with BrS (mean age 47 years; 111 men) who were treated with an ICD. The patients were classified into 3 groups based on symptoms at implantation: asymptomatic (35 patients); LOC (46 patients); and ventricular tachyarrhythmia (VTA) (31 patients). We evaluated the incidence and cause of LOC during long-term follow-up after ICD implantation. Results During mean follow-up of 12.2 years, 41 patients (37%) experienced LOC after ICD implantation. Arrhythmic LOC occurred in 5 asymptomatic patients, 14 LOC patients, and 16 patients with VTA. Nonarrhythmic LOC, similar to the initial episode, occurred after ICD implantation in 6 patients with prior LOC (2 with neurally mediated syncope and 4 with epilepsy). Most epileptic patients experienced LOC during rest or sleeping, and did not show an abnormal encephalogram during initial evaluation of the LOC episodes. Conclusion After ICD implantation, 13% of patients had nonarrhythmic LOC similar to the initial episode. Accurate classification of LOC based on a detailed medical history is important for risk stratification, although distinguishing arrhythmic LOC from epilepsy-related LOC episodes can be challenging depending on the circumstances and characteristics of the LOC event.
Collapse
Affiliation(s)
- Saori Asada
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Morita
- Department of Cardiovascular Therapeutics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tomofumi Mizuno
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuro Masuda
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akira Ueoka
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masakazu Miyamoto
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Kawada
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Nakagawa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nobuhiro Nishii
- Department of Cardiovascular Therapeutics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
32
|
Antonucci F, Bozzi Y. Action of Botulinum Neurotoxin E Type in Experimental Epilepsies. Toxins (Basel) 2023; 15:550. [PMID: 37755976 PMCID: PMC10536604 DOI: 10.3390/toxins15090550] [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: 07/21/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are zinc endopeptidases produced by the Clostridium genus of anerobic bacteria, largely known for their ability to cleave synaptic proteins, leading to neuromuscular paralysis. In the central nervous system, BoNTs are known to block the release of glutamate neurotransmitter, and for this reason, researchers explored the possible therapeutic action in disorders characterized by neuronal hyperactivity, such as epilepsy. Thus, using multidisciplinary approaches and models of experimental epilepsy, we investigated the pharmacological potential of BoNT/E serotype. In this review, written in memory of Prof. Matteo Caleo, a pioneer in these studies, we go back over the hypotheses and experimental approaches that led us to the conclusion that intrahippocampal administration of BoNT/E (i) displays anticonvulsant effects if prophylactically delivered in a model of acute generalized seizures; (ii) does not have any antiepileptogenic action after the induction of status epilepticus; (iii) reduces frequency of spontaneous seizures in a model of recurrent seizures if delivered during the chronic phase but in a transient manner. Indeed, the control on spontaneous seizures stops when BoNT/E effects are off (few days), thus limiting its pharmacological potential in humans.
Collapse
Affiliation(s)
- Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, via Fratelli Cervi 93, 20054 Milan, Italy
- CNR Institute of Neuroscience, via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
| | - Yuri Bozzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
- CNR Institute of Neuroscience, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
33
|
Liu Y, Bassetto CAZ, Pinto BI, Bezanilla F. A mechanistic reinterpretation of fast inactivation in voltage-gated Na + channels. Nat Commun 2023; 14:5072. [PMID: 37604801 PMCID: PMC10442390 DOI: 10.1038/s41467-023-40514-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023] Open
Abstract
The hinged-lid model was long accepted as the canonical model for fast inactivation in Nav channels. It predicts that the hydrophobic IFM motif acts intracellularly as the gating particle that binds and occludes the pore during fast inactivation. However, the observation in recent high-resolution structures that the bound IFM motif is located far from the pore, contradicts this preconception. Here, we provide a mechanistic reinterpretation of fast inactivation based on structural analysis and ionic/gating current measurements. We demonstrate that in Nav1.4 the final inactivation gate is comprised of two hydrophobic rings at the bottom of S6 helices. These rings function in series and close downstream of IFM binding. Reducing the volume of the sidechain in both rings leads to a partially conductive, leaky inactivated state and decreases the selectivity for Na+ ion. Altogether, we present an alternative molecular framework to describe fast inactivation.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Bernardo I Pinto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
34
|
Miao P, Zhu X, Jin W, Yu L, Li Y, Wang Y, Su Q, Xu S, Wang S, Feng J. Efficacy of perampanel in pediatric epilepsy with known and presumed genetic etiology. Ann Clin Transl Neurol 2023; 10:1374-1382. [PMID: 37329172 PMCID: PMC10424658 DOI: 10.1002/acn3.51828] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 06/18/2023] Open
Abstract
OBJECTIVE The efficacy of perampanel (PER) in pediatric epilepsy with specific etiologies has not been well established. Here, we investigated outcome and predictors of PER treatment in a pediatric cohort with known and presumed genetic etiology. METHODS We included pediatric patients with potential genetic epilepsy who received PER treatment and underwent whole-exome sequencing (WES) from January 2020 to September 2021. All patients were followed up for >12 months. RESULTS A total of 124 patients were included. Overall response rates were 51.6% and 49.6% at 6 months and 12 months, respectively. Pathogenic or likely pathogenic variants in 27 multiple genes were detected among 58 patients (46.8%) by WES. On performing multivariate logistic regression analysis, only developmental delay (OR = 0.406, P = 0.042) was a negative predictor of treatment response. However, the seizure onset age, positive WES results, and number of ASMs before PER administration were not significantly. Thirteen carriers with variants in the SCN1A gene showed a better response compared to eight patients with other sodium channels (P = 0.007), and to the other 45 patients with positive WES results (OR = 7.124, 95% CI = 1.306-38.860, P = 0.023). Adverse events were only reported in 23 patients, the most common being emotional problems. INTERPRETATION PER is safe and efficacious in pediatric patients with known and presumed genetic etiology. The response rate is comparable to that reported in other pediatric populations, and lower among those with developmental delay. A gene-specific response to PER is found along with better efficacy links to pathogenic variants in the SCN1A gene.
Collapse
Affiliation(s)
- Pu Miao
- Department of PediatricsSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Xueying Zhu
- Department of PediatricsJinhua Lanxi People's HospitalJinhuaZhejiang321000China
| | - Wenqin Jin
- Department of PediatricsSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Lingyan Yu
- Department of PharmacySecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yanfang Li
- Department of PediatricsSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Ye Wang
- Department of PediatricsSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Qunyan Su
- Department of PediatricsTaizhou Woman and Children's HospitalTaizhou318000China
| | - Sha Xu
- Department of Neurology, Epilepsy CenterSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Shuang Wang
- Department of Neurology, Epilepsy CenterSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| | - Jianhua Feng
- Department of PediatricsSecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou310009China
| |
Collapse
|
35
|
Paruch K, Kaproń B, Łuszczki JJ, Paneth A, Plech T. Effect of Linker Elongation on the VGSC Affinity and Anticonvulsant Activity among 4-Alkyl-5-aryl-1,2,4-triazole-3-thione Derivatives. Molecules 2023; 28:5287. [PMID: 37446948 DOI: 10.3390/molecules28135287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
The main aim of the current project was to investigate the effect of the linker size in 4-alkyl-5-aryl-1,2,4-triazole-3-thione derivatives, known as a group of antiepileptic drug candidates, on their affinity towards voltage-gated sodium channels (VGSCs). The rationale of the study was based both on the SAR observations and docking simulations of the interactions between the designed ligands and the binding site of human VGSC. HYDE docking scores, which describe hydrogen bonding, desolvation, and hydrophobic effects, obtained for 5-[(3-chlorophenyl)ethyl]-4-butyl/hexyl-1,2,4-triazole-3-thiones, justified their beneficial sodium channel blocking activity. The results of docking simulations were verified using a radioligand binding assay with [3H]batrachotoxin. Unexpectedly, although the investigated triazole-based compounds acted as VGSC ligands, their affinities were lower than those of the respective analogs containing shorter alkyl linkers. Since numerous sodium channel blockers are recognized as antiepileptic agents, the obtained 1,2,4-triazole derivatives were examined for antiepileptic potential using an experimental model of tonic-clonic seizures in mice. Median effective doses (ED50) of the compounds examined in MES test reached 96.6 ± 14.8 mg/kg, while their median toxic doses (TD50), obtained in the rotarod test, were even as high as 710.5 ± 47.4 mg/kg.
Collapse
Affiliation(s)
- Kinga Paruch
- Department of Pharmacology, Faculty of Health Sciences, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland
| | - Barbara Kaproń
- Department of Clinical Genetics, Faculty of Medicine, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Occupational Medicine, Faculty of Medical Sciences, Medical University of Lublin, Jaczewskiego 8B, 20-090 Lublin, Poland
| | - Agata Paneth
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4A, 20-059 Lublin, Poland
| | - Tomasz Plech
- Department of Pharmacology, Faculty of Health Sciences, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland
| |
Collapse
|
36
|
Chapman DP, Vicini S, Burns MP, Evans R. Single Neuron Modeling Identifies Potassium Channel Modulation as Potential Target for Repetitive Head Impacts. Neuroinformatics 2023; 21:501-516. [PMID: 37294503 PMCID: PMC10833395 DOI: 10.1007/s12021-023-09633-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Traumatic brain injury (TBI) and repetitive head impacts can result in a wide range of neurological symptoms. Despite being the most common neurological disorder in the world, repeat head impacts and TBI do not have any FDA-approved treatments. Single neuron modeling allows researchers to extrapolate cellular changes in individual neurons based on experimental data. We recently characterized a model of high frequency head impact (HFHI) with a phenotype of cognitive deficits associated with decreases in neuronal excitability of CA1 neurons and synaptic changes. While the synaptic changes have been interrogated in vivo, the cause and potential therapeutic targets of hypoexcitability following repetitive head impacts are unknown. Here, we generated in silico models of CA1 pyramidal neurons from current clamp data of control mice and mice that sustained HFHI. We use a directed evolution algorithm with a crowding penalty to generate a large and unbiased population of plausible models for each group that approximated the experimental features. The HFHI neuron model population showed decreased voltage gated sodium conductance and a general increase in potassium channel conductance. We used partial least squares regression analysis to identify combinations of channels that may account for CA1 hypoexcitability after HFHI. The hypoexcitability phenotype in models was linked to A- and M-type potassium channels in combination, but not by any single channel correlations. We provide an open access set of CA1 pyramidal neuron models for both control and HFHI conditions that can be used to predict the effects of pharmacological interventions in TBI models.
Collapse
Affiliation(s)
- Daniel P Chapman
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mark P Burns
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| | - Rebekah Evans
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
37
|
Bouzroud W, Tazzite A, Boussakri I, Gazzaz B, Dehbi H. A novel SCN8A variant of unknown significance in pediatric epilepsy: a case report. J Int Med Res 2023; 51:3000605231187931. [PMID: 37498161 PMCID: PMC10387795 DOI: 10.1177/03000605231187931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023] Open
Abstract
Variants in SCN8A are associated with several diseases, including developmental and epileptic encephalopathy, intermediate epilepsy or mild-to-moderate developmental and epileptic encephalopathy, self-limited familial infantile epilepsy, neurodevelopmental delays with generalized epilepsy, neurodevelopmental disorder without epilepsy, hypotonia, and movement disorders. Herein, we report an 8-year-old Moroccan boy with intermediate epilepsy of unknown origin, intellectual disability, autism spectrum disorder, and hyperactivity. The patient presented a normal 46, XY karyotype and a normal comparative genomic hybridization profile. Whole-exome sequencing was performed, and heterozygous variants were identified in KCNK4 and SCN8A. The SCN8A variant [c.4499C > T (p.Pro1500Leu)] was also detected in the healthy mother and was classified as a variant of uncertain clinical significance. This variant occurs in a highly conserved domain, which may affect the function of the encoded protein. More studies are needed to confirm the pathogenicity of this novel variant to establish the effective care, management, and genetic counselling of affected individuals.
Collapse
Affiliation(s)
- Wafaa Bouzroud
- Medical Genetics Laboratory, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Amal Tazzite
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ikhlass Boussakri
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Bouchaïb Gazzaz
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
- Genetics Analysis Institute, Royal Gendarmerie, Rabat, Morocco
| | - Hind Dehbi
- Medical Genetics Laboratory, Ibn Rochd University Hospital, Casablanca, Morocco
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
38
|
Zhu J, Lu J, Shen X, He Y, Xia H, Li W, Guo H, Zhang J, Fan X. SCN1A Polymorphisms and Haplotypes Are Associated With Valproic Acid Treatment Outcomes in Chinese Children With Epilepsy. Pediatr Neurol 2023; 146:55-64. [PMID: 37451178 DOI: 10.1016/j.pediatrneurol.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/20/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Sodium channel genes, especially SCN1A, were reported to play an important role in the treatment outcomes of antiseizure medications. The aim of this study was to explore the association of SCN1A polymorphisms with efficacy and adverse drug reactions (ADRs) related to valproic acid (VPA) among Chinese children with epilepsy. METHODS A total of 126 children with epilepsy treated with VPA for at least 12 months were enrolled in this study. Three single nucleotide polymorphisms (SNPs) of SCN1A including rs2298771, rs10167228, and rs3812718 were genotyped using Sequenom MassArray system. Bioinformatics tools were used to explore the potential targets and pathways of SCN1A in VPA-related ADRs. RESULTS The three SNPs in this study were found to be closely associated with treatment outcomes for VPA. Carriers of SCN1A rs3812718 TT genotype tended to be seizure-free with VPA treatment (P = 0.007). AA genotype of rs10167228 and TT genotype of rs2298771 might be protective factors for weight gain induced by VPA, whereas TA genotype of rs10167228 and CT genotype of rs2298771 increased the risk. TAT haplotype carriers were found to respond better to VPA treatment (P = 0.017), whereas CTC haplotype might be a risk factor for VPA-induced weight gain (P = 0.035). Bioinformatics analysis suggested that SCN1A might play a role in VPA-induced weight gain by regulating gated channel activity and GABAergic synapse pathway. CONCLUSION This study revealed that SCN1A rs2298771, rs10167228, and rs3812718 polymorphisms and haplotypes might affect the treatment outcomes of VPA in Chinese children with epilepsy.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China; Department of Clinical Pharmacology, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jieluan Lu
- Department of Clinical Pharmacology, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Xianhuan Shen
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China; Department of Clinical Pharmacology, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Yaodong He
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China; Department of Clinical Pharmacology, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Hanbing Xia
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Wenzhou Li
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Huijuan Guo
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Jianping Zhang
- Department of Clinical Pharmacology, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| | - Xiaomei Fan
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China.
| |
Collapse
|
39
|
Hung TY, Wu SN, Huang CW. Concerted suppressive effects of carisbamate, an anti-epileptic alkyl-carbamate drug, on voltage-gated Na + and hyperpolarization-activated cation currents. Front Cell Neurosci 2023; 17:1159067. [PMID: 37293624 PMCID: PMC10244622 DOI: 10.3389/fncel.2023.1159067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Carisbamate (CRS, RWJ-333369) is a new anti-seizure medication. It remains unclear whether and how CRS can perturb the magnitude and/or gating kinetics of membrane ionic currents, despite a few reports demonstrating its ability to suppress voltage-gated Na+ currents. In this study, we observed a set of whole-cell current recordings and found that CRS effectively suppressed the voltage-gated Na+ (INa) and hyperpolarization-activated cation currents (Ih) intrinsically in electrically excitable cells (GH3 cells). The effective IC50 values of CRS for the differential suppression of transient (INa(T)) and late INa (INa(L)) were 56.4 and 11.4 μM, respectively. However, CRS strongly decreased the strength (i.e., Δarea) of the nonlinear window component of INa (INa(W)), which was activated by a short ascending ramp voltage (Vramp); the subsequent addition of deltamethrin (DLT, 10 μM) counteracted the ability of CRS (100 μM, continuous exposure) to suppress INa(W). CRS strikingly decreased the decay time constant of INa(T) evoked during pulse train stimulation; however, the addition of telmisartan (10 μM) effectively attenuated the CRS (30 μM, continuous exposure)-mediated decrease in the decay time constant of the current. During continued exposure to deltamethrin (10 μM), known to be a pyrethroid insecticide, the addition of CRS resulted in differential suppression of the amplitudes of INa(T) and INa(L). The amplitude of Ih activated by a 2-s membrane hyperpolarization was diminished by CRS in a concentration-dependent manner, with an IC50 value of 38 μM. For Ih, CRS altered the steady-state I-V relationship and attenuated the strength of voltage-dependent hysteresis (Hys(V)) activated by an inverted isosceles-triangular Vramp. Moreover, the addition of oxaliplatin effectively reversed the CRS-mediated suppression of Hys(V). The predicted docking interaction between CRS and with a model of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel or between CRS and the hNaV1.7 channel reflects the ability of CRS to bind to amino acid residues in HCN or hNaV1.7 channel via hydrogen bonds and hydrophobic interactions. These findings reveal the propensity of CRS to modify INa(T) and INa(L) differentially and to effectively suppress the magnitude of Ih. INa and Ih are thus potential targets of the actions of CRS in terms of modulating cellular excitability.
Collapse
Affiliation(s)
- Te-Yu Hung
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- College of Medicine, Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
40
|
Liu Y, Bassetto CAZ, Pinto BI, Bezanilla F. A Mechanistic Reinterpretation of Fast Inactivation in Voltage-Gated Na+ Channels. RESEARCH SQUARE 2023:rs.3.rs-2924505. [PMID: 37292679 PMCID: PMC10246267 DOI: 10.21203/rs.3.rs-2924505/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The hinged-lid model is long accepted as the canonical model for fast inactivation in Nav channels. It predicts that the hydrophobic IFM motif acts intracellularly as the gating particle that binds and occludes the pore during fast inactivation. However, the observation in recent high-resolution structures that the bound IFM motif locates far from the pore, contradicts this preconception. Here, we provide a mechanistic reinterpretation of fast inactivation based on structural analysis and ionic/gating current measurements. We demonstrate that in Nav1.4 the final inactivation gate is comprised of two hydrophobic rings at the bottom of S6 helices. These rings function in series and close downstream of IFM binding. Reducing the volume of the sidechain in both rings leads to a partially conductive "leaky" inactivated state and decreases the selectivity for Na + ion. Altogether, we present an alternative molecular framework to describe fast inactivation.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Bernardo I Pinto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Centro Interdisciplinario de Neurociencias de Valparaiso, Valparaiso, Chile
| |
Collapse
|
41
|
Bali A, Schaefer SP, Trier I, Zhang AL, Kabeche L, Paulsen CE. Molecular mechanism of hyperactivation conferred by a truncation of TRPA1. Nat Commun 2023; 14:2867. [PMID: 37208332 PMCID: PMC10199097 DOI: 10.1038/s41467-023-38542-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/08/2023] [Indexed: 05/21/2023] Open
Abstract
A drastic TRPA1 mutant (R919*) identified in CRAMPT syndrome patients has not been mechanistically characterized. Here, we show that the R919* mutant confers hyperactivity when co-expressed with wild type (WT) TRPA1. Using functional and biochemical assays, we reveal that the R919* mutant co-assembles with WT TRPA1 subunits into heteromeric channels in heterologous cells that are functional at the plasma membrane. The R919* mutant hyperactivates channels by enhancing agonist sensitivity and calcium permeability, which could account for the observed neuronal hypersensitivity-hyperexcitability symptoms. We postulate that R919* TRPA1 subunits contribute to heteromeric channel sensitization by altering pore architecture and lowering energetic barriers to channel activation contributed by the missing regions. Our results expand the physiological impact of nonsense mutations, reveal a genetically tractable mechanism for selective channel sensitization, uncover insights into the process of TRPA1 gating, and provide an impetus for genetic analysis of patients with CRAMPT or other stochastic pain syndromes.
Collapse
Affiliation(s)
- Avnika Bali
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Samantha P Schaefer
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Isabelle Trier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Alice L Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Candice E Paulsen
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA.
| |
Collapse
|
42
|
Barbieri R, Nizzari M, Zanardi I, Pusch M, Gavazzo P. Voltage-Gated Sodium Channel Dysfunctions in Neurological Disorders. Life (Basel) 2023; 13:life13051191. [PMID: 37240836 DOI: 10.3390/life13051191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
The pore-forming subunits (α subunits) of voltage-gated sodium channels (VGSC) are encoded in humans by a family of nine highly conserved genes. Among them, SCN1A, SCN2A, SCN3A, and SCN8A are primarily expressed in the central nervous system. The encoded proteins Nav1.1, Nav1.2, Nav1.3, and Nav1.6, respectively, are important players in the initiation and propagation of action potentials and in turn of the neural network activity. In the context of neurological diseases, mutations in the genes encoding Nav1.1, 1.2, 1.3 and 1.6 are responsible for many forms of genetic epilepsy and for Nav1.1 also of hemiplegic migraine. Several pharmacological therapeutic approaches targeting these channels are used or are under study. Mutations of genes encoding VGSCs are also involved in autism and in different types of even severe intellectual disability (ID). It is conceivable that in these conditions their dysfunction could indirectly cause a certain level of neurodegenerative processes; however, so far, these mechanisms have not been deeply investigated. Conversely, VGSCs seem to have a modulatory role in the most common neurodegenerative diseases such as Alzheimer's, where SCN8A expression has been shown to be negatively correlated with disease severity.
Collapse
Affiliation(s)
| | - Mario Nizzari
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Ilaria Zanardi
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Michael Pusch
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Paola Gavazzo
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| |
Collapse
|
43
|
Liu Y, Bassetto CAZ, Pinto BI, Bezanilla F. A Mechanistic Reinterpretation of Fast Inactivation in Voltage-Gated Na + Channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.27.538555. [PMID: 37162849 PMCID: PMC10168311 DOI: 10.1101/2023.04.27.538555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Fast Inactivation in voltage-gated Na + channels plays essential roles in numerous physiological functions. The canonical hinged-lid model has long predicted that a hydrophobic motif in the DIII-DIV linker (IFM) acts as the gating particle that occludes the permeation pathway during fast inactivation. However, the fact that the IFM motif is located far from the pore in recent high-resolution structures of Nav + channels contradicts this status quo model. The precise molecular determinants of fast inactivation gate once again, become an open question. Here, we provide a mechanistic reinterpretation of fast inactivation based on ionic and gating current data. In Nav1.4 the actual inactivation gate is comprised of two hydrophobic rings at the bottom of S6. These function in series and closing once the IFM motif binds. Reducing the volume of the sidechain in both rings led to a partially conductive inactivated state. Our experiments also point to a previously overlooked coupling pathway between the bottom of S6 and the selectivity filter.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Bernardo I Pinto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Centro Interdisciplinario de Neurociencias de Valparaiso, Valparaiso, Chile
| |
Collapse
|
44
|
Nakatani Y, Ishikawa K, Aoki Y, Shimooki T, Yamamoto N, Amano T. Inhibitory effect of atomoxetine on Nav1.2 voltage-gated sodium channel currents. Pharmacol Rep 2023; 75:746-752. [PMID: 36914846 DOI: 10.1007/s43440-023-00477-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Atomoxetine (ATX), a norepinephrine reuptake inhibitor (NRI), is used to attenuate the symptoms of Attention Deficit/Hyperactivity Disorder (AD/HD) by increasing neurotransmitter concentrations at the synaptic cleft. Although Nav1.2 voltage-gated sodium channels (VGSCs) are thought to play a role in monoamine transmitter release in the synaptic junction, it is unclear how atomoxetine affects Nav1.2 VGSCs. METHODS In this study, we investigated the effect of ATX on Nav1.2 VGSC-transfected HEK293 cells with the whole-patch clamp technique. RESULTS Nav1.2 VGSC current decreased by 51.15 ± 12.75% under treatment with 50 µM ATX in the resting state (holding membrane potential at - 80 mV). The IC50 of ATX against Nav1.2 VGSC current was 45.57 µM. The activation/inactivation curve of Nav1.2 VGSC currents was shifted toward hyperpolarization by 50 µM ATX. In addition, the inhibitory effect of ATX increased with membrane depolarization (holding membrane potential at - 50 mV) and its IC50 was 10.16 µM. Moreover, ATX showed the time-dependent interaction in the inactivation state. CONCLUSION These findings suggest that ATX interacts with Nav1.2 VGSCs producing the inhibition of current and the modification of kinetic properties in the state-dependent manner.
Collapse
Affiliation(s)
- Yoshihiko Nakatani
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan.
| | - Kanami Ishikawa
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan
| | - Yuko Aoki
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan
| | - Takahiro Shimooki
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan
| | - Naoki Yamamoto
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan.,Center for Basic Medical Research, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan.,Laboratory of Neurobiology, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-Shi, Tokyo, 192-0397, Japan
| | - Taku Amano
- Tochigi Prefectural Okamotodai Hospital, 2162 Shimookamotomachi, Utsunomiya, Tochigi, 329-1104, Japan
| |
Collapse
|
45
|
Menezes LFS, Maranhão MM, Tibery DV, de Souza ACB, da Mata DO, Campos LA, Souza AA, Freitas SMD, Schwartz EF. Ts17, a Tityus serrulatus β-toxin structurally related to α-scorpion toxins. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184057. [PMID: 36240866 DOI: 10.1016/j.bbamem.2022.184057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022]
Abstract
Ts17 was purified from the venom of the scorpion Tityus serrulatus, the most dangerous scorpion species in Brazil. The activity on Nav1.1-Nav1.7 channels was electrophysiologically characterized by patch-clamp technique. Ts17 amino acid sequence indicated high similarity to alpha-scorpion toxins; however, it presented beta-toxin activity, altering the kinetics of the Na+-channels. The most affected subtypes during activation (with and without prepulse) and inactivation phases were Nav1.2 and Nav1.5, respectively. For recovery from inactivation, the most affected voltage-gated sodium channel was Nav1.5. Circular dichroism spectra showed that Ts17 presents mainly β-sheet and unordered structures at all analyzed pHs, and the maximum value of α-helix was found at pH 4.0 (13.3 %). Based on the results, Ts17 might be used as a template to develop a new cardiac drug. Key contribution Purification of Ts17 from Tityus serrulatus, electrophysiological characterization of Ts17 on voltage-gated sodium channel subtypes, β-toxin classification.
Collapse
Affiliation(s)
- Luis Felipe Santos Menezes
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Mariza Mendanha Maranhão
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Adolfo Carlos Barros de Souza
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Daniel Oliveira da Mata
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Leandro Ambrósio Campos
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil; Instituto de Ciências da Saúde, Universidade Paulista, Brasília 70390-130, Distrito Federal, Brazil
| | - Amanda Araújo Souza
- Brazilian Biosciences National Laboratory (LNBio), National Center for Research in Energy and Materials (CNPEM), Campinas 13083-970, São Paulo, Brazil
| | - Sonia Maria de Freitas
- Laboratório de Biofísica Molecular, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil.
| |
Collapse
|
46
|
Guo QB, Zhan L, Xu HY, Gao ZB, Zheng YM. SCN8A epileptic encephalopathy mutations display a gain-of-function phenotype and divergent sensitivity to antiepileptic drugs. Acta Pharmacol Sin 2022; 43:3139-3148. [PMID: 35902765 PMCID: PMC9712530 DOI: 10.1038/s41401-022-00955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/05/2022] [Indexed: 11/09/2022] Open
Abstract
De novo missense mutations in SCN8A gene encoding voltage-gated sodium channel NaV1.6 are linked to a severe form of early infantile epileptic encephalopathy named early infantile epileptic encephalopathy type13 (EIEE13). The majority of the patients with EIEE13 does not respond favorably to the antiepileptic drugs (AEDs) in clinic and has a significantly increased risk of death. Although more than 60 EIEE13-associated mutations have been discovered, only few mutations have been functionally analyzed. In this study we investigated the functional influences of mutations N1466T and N1466K, two EIEE13-associated mutations located in the inactivation gate, on sodium channel properties. Sodium currents were recorded from CHO cells expressing the mutant and wide-type (WT) channels using the whole-cell patch-clamp technique. We found that, in comparison with WT channels, both the mutant channels exhibited increased window currents, persistent currents (INaP) and ramp currents, suggesting that N1466T and N1466K were gain-of-function (GoF) mutations. Sodium channel inhibition is one common mechanism of currently available AEDs, in which topiramate (TPM) was effective in controlling seizures of patients carrying either of the two mutations. We found that TPM (100 µM) preferentially inhibited INaP and ramp currents but did not affect transient currents (INaT) mediated by N1466T or N1466K. Among the other 6 sodium channel-inhibiting AEDs tested, phenytoin and carbamazepine displayed greater efficacy than TPM in suppressing both INaP and ramp currents. Functional characterization of mutants N1466T and N1466K is beneficial for understanding the pathogenesis of EIEE13. The divergent effects of sodium channel-inhibiting AEDs on INaP and ramp currents provide insight into the development of therapeutic strategies for the N1466T and N1466K-associated EIEE13.
Collapse
Affiliation(s)
- Qian-Bei Guo
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Zhan
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai-Yan Xu
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhao-Bing Gao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China.
| | - Yue-Ming Zheng
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
47
|
Makridis KL, Friedo AL, Kellinghaus C, Losch FP, Schmitz B, Boßelmann C, Kaindl AM. Successful treatment of adult Dravet syndrome patients with cenobamate. Epilepsia 2022; 63:e164-e171. [PMID: 36176237 DOI: 10.1111/epi.17427] [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: 05/17/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 01/11/2023]
Abstract
Dravet syndrome (DS) is a rare, drug-resistant, severe developmental and epileptic encephalopathy caused by pathogenic variants in the α subunit of the voltage-gated sodium channel gene SCN1A. Hyperexcitability in DS results from loss of function in inhibitory interneurons. Thus sodium channel blockers are usually contraindicated in patients with DS as they may lead to disease aggravation. Cenobamate (CNB) is a novel antiseizure medication (ASM) with promising rates of seizure freedom in patients with focal-onset, drug-resistant epilepsy. CNB blocks persistent sodium currents by promoting the inactive states of sodium channels. In a multi-center study, we analyzed retrospectively the effect of an add-on therapy of CNB in adult patients with DS. We report four adult patients with DS in whom the use of CNB resulted in a significant seizure reduction of more than 80%, with a follow-up of up to 542 days. CNB was the first drug in these patients that resulted in a long-lasting and significant seizure reduction. No severe adverse events occurred. We highlight CNB as an ASM that may lead to a clinically meaningful reduction of seizure frequency in adult patients with DS. It is unclear, however, if all patients with DS benefit, requiring further investigation and functional experiments.
Collapse
Affiliation(s)
- Konstantin L Makridis
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Cell and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Epilepsy Center for Children and Adolescents, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna-Lena Friedo
- Epilepsy Center Berlin-Brandenburg, Epilepsieklinik Tabor, Bernau, Germany
| | | | | | - Bettina Schmitz
- Department of Neurology, Vivantes Humboldt-Klinikum, Berlin, Germany
| | - Christian Boßelmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Cell and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Epilepsy Center for Children and Adolescents, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
48
|
Morato EO, Knight B, Nair LS. Transcriptional profiling of neuronal ion channels in dorsal root ganglion-derived immortal cell line (F-11) under different culture conditions. IN VITRO MODELS 2022; 1:385-395. [PMID: 38107764 PMCID: PMC10723754 DOI: 10.1007/s44164-022-00036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 12/19/2023]
Abstract
Pathological pain is a prevalent condition that affects majority of adults with a variety of underlying disease conditions. Current available pharmacological pain treatments have several negative and potentially life-threatening side effects associated with their long-term use. Due to the heterogeneity of pain perception and the diversity of neuronal mechanisms that contribute to pain, high-throughput screening of small molecules that may have underlying analgesic properties is essential for identifying new analgesic treatments that are both effective and safe. The F-11 hybrid immortalized cell line is one of the currently available dorsal root ganglion (DRG) cell lines used for drug screening. While F-11 cells are commonly used as analogs to primary DRG sensory neurons, they differ significantly in physiological properties. The present study investigated the impact of differentiation protocols on the expression of mature neuron ion channels and receptors in F-11 cells. Using a customized gene array of more than eighty neuronal ion channels and receptors including voltage-gated ion channels, transient receptor potential channels, and cannabinoid receptors, we assessed the following groups: control F-11 cells; F-11 cells cultured under different culture conditions, and murine DRG tissue. The expression profiles of majority of the investigated ion channels and receptors in F-11 cells were found to be lower compared to primary mouse DRG neurons. F-11 cells cultured under low serum (LSM) conditions had increased expression of several investigated targets including voltage-gated ion channels and cannabinoid receptors when compared to control F-11 cells. The study showed that the culture conditions significantly modulated the transcriptional expression of studied ion channels and receptors, and that long-term culture (21 days) may adversely affect the expression of many of the studied targets.
Collapse
Affiliation(s)
- Erick Orozco Morato
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Brittany Knight
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Lakshmi S. Nair
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Biomedical Engineering, Department of Material Science and Engineering, Institute of Material Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
49
|
Omer H, Omer MH, Alyousef AR, Alzammam AM, Ahmad O, Alanazi HA. Unmasking of Brugada syndrome by lamotrigine in a patient with pre-existing epilepsy: A case report with review of the literature. Front Cardiovasc Med 2022; 9:1005952. [PMID: 36407465 PMCID: PMC9673589 DOI: 10.3389/fcvm.2022.1005952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/10/2022] [Indexed: 11/26/2022] Open
Abstract
Brugada syndrome is an inherited cardiac channelopathy arising from mutations in voltage-gated cardiac sodium channels. Idiopathic epilepsy portrays a coalescent underlying pathophysiological mechanism pertaining to the premature excitation of neuronal voltage-gated ion channels resulting in the disruption of presynaptic neurons and the unregulated release of excitatory neurotransmitters. The coexistence of epilepsy and Brugada syndrome may be explained by mutations in voltage-gated ion channels, which are coexpressed in cardiac and neural tissue. Moreover, the incidence of sudden unexpected death in epilepsy has been associated with malignant cardiac arrhythmias in the presence of mutations in voltage-gated ion channels. Lamotrigine is an antiepileptic drug that inhibits neuronal voltage-gated sodium channels, thus stabilizing neural impulse propagation and controlling seizure activity in the brain. However, lamotrigine has been shown to inhibit cardiac voltage-gated sodium channels resulting in a potential arrhythmogenic effect and the ability to unmask Brugada syndrome in genetically susceptible individuals. We are reporting a case of a 27-year-old male patient with a background of presumed idiopathic epilepsy who was initiated on lamotrigine therapy resulting in the unmasking of Brugada syndrome and the onset of syncopal episodes. This case provides further evidence for the arrhythmogenic capacity of lamotrigine and highlights the relationship between epilepsy and Brugada syndrome. In this report, we aim to review the current literature regarding the associations between epilepsy and Brugada syndrome and the impact of lamotrigine therapy on such patients.
Collapse
Affiliation(s)
- Hafiz Omer
- Department of Adult Cardiology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- *Correspondence: Hafiz Omer,
| | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | - Ali M. Alzammam
- Department of Internal Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Omar Ahmad
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Haitham A. Alanazi
- Department of Adult Cardiology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
50
|
Lu Q, Wang YY, Chen HM, Wang QH, Yang XY, Zou LP. A rise in saliva and urine pH in children with SCN1A-related epilepsy: An exploratory prospective controlled study. Front Neurol 2022; 13:982050. [PMID: 36237607 PMCID: PMC9552845 DOI: 10.3389/fneur.2022.982050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Objective SCN1A, encoding the alpha 1 subunit of the sodium channel, is associated with a range of related epilepsy. This study aims to assess saliva and urine pH in children with SCN1A-related epilepsy. Methods A prospective controlled observational study with a 1:1 ratio was conducted on seven patients with SCN1A-related epilepsy and seven healthy children of the same family, gender, and age but without a history of seizures. The pH of saliva and urine was measured by pH test paper. Parents of patients with epilepsy recorded seizures to compare the relationship between pH and seizures. Results The fourteen participants were all males, aged 1 to 14 years. Seven patients had different pathogenic SCN1A variants. The pH of saliva and urine was monitored for 21–95 days. The pH of saliva and urine was higher in patients with SCN1A-related epilepsy than in the healthy group. The urine pH in Dravet syndrome patients was high compared with other epilepsy patients. The urine pH in patients with seizures was higher than that in patients without seizures, which occurred during the study. Conclusions The pH of saliva and urine was chronically high in patients with SCN1A-related epilepsy, and urine pH was higher in patients with seizures and with Dravet syndrome.
Collapse
Affiliation(s)
- Qian Lu
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang-Yang Wang
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui-Min Chen
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiu-Hong Wang
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Yan Yang
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li-Ping Zou
- Senior Department of Pediatrics, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Beijing Institute for Brain Disorders, Center for Brain Disorders Research, Capital Medical University, Beijing, China
- *Correspondence: Li-Ping Zou
| |
Collapse
|