1
|
Edmond MA, Hinojo-Perez A, Efrem M, Yi-Chun L, Shams I, Hayoz S, de la Cruz A, Perez Rodriguez ME, Diaz-Solares M, Dykxhoorn DM, Luo YL, Barro-Soria R. Lipophilic compounds restore function to neurodevelopmental-associated KCNQ3 mutations. Commun Biol 2024; 7:1181. [PMID: 39300259 DOI: 10.1038/s42003-024-06873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
A major driver of neuronal hyperexcitability is dysfunction of K+ channels, including voltage-gated KCNQ2/3 channels. Their hyperpolarized midpoint of activation and slow activation and deactivation kinetics produce a current that regulates membrane potential and impedes repetitive firing. Inherited mutations in KCNQ2 and KCNQ3 are linked to a wide spectrum of neurodevelopmental disorders (NDDs), ranging from benign familial neonatal seizures to severe epileptic encephalopathies and autism spectrum disorders. However, the impact of these variants on the molecular mechanisms underlying KCNQ3 channel function remains poorly understood and existing treatments have significant side effects. Here, we use voltage clamp fluorometry, molecular dynamic simulations, and electrophysiology to investigate NDD-associated variants in KCNQ3 channels. We identified two distinctive mechanisms by which loss- and gain-of function NDD-associated mutations in KCNQ3 affect channel gating: one directly affects S4 movement while the other changes S4-to-pore coupling. MD simulations and electrophysiology revealed that polyunsaturated fatty acids (PUFAs) primarily target the voltage-sensing domain in its activated conformation and form a weaker interaction with the channel's pore. Consistently, two such compounds yielded partial and complete functional restoration in R227Q- and R236C-containing channels, respectively. Our results reveal the potential of PUFAs to be developed into therapies for diverse KCNQ3-based channelopathies.
Collapse
Affiliation(s)
- Michaela A Edmond
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Texas A&M University Health Science Center, Department of Neuroscience & Experimental Therapeutics, Bryan, USA
| | - Andy Hinojo-Perez
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mekedlawit Efrem
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Lin Yi-Chun
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Iqra Shams
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sebastien Hayoz
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Physiology, University of Arizona, Tucson, USA
| | - Alicia de la Cruz
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Linkoping University, Department of Biomedical and Clinical Sciences (BKV), Linkoping, Sweden
| | | | - Maykelis Diaz-Solares
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Yun Lyna Luo
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Rene Barro-Soria
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
2
|
Zhang Y, Xue Y, Ma Y, Du X, Lu B, Wang Y, Yan Z. Improved classification and pathogenicity assessment by comprehensive functional studies in a large data set of KCNQ2 variants. Life Sci 2024; 339:122378. [PMID: 38142737 DOI: 10.1016/j.lfs.2023.122378] [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: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
AIMS The paucity of functional annotations on hundreds of KCNQ2 variants impedes the diagnosis and treatment of KCNQ2-related disorders. The aims of this work were to determine the functional properties of 331 clinical KCNQ2 variants, interpreted the pathogenicity of 331 variants using functional data,and explored the association between homomeric channel functions and phenotypes. MAIN METHODS We collected 145 KCNQ2 variants from 232 epilepsy patients and 186 KCNQ2 missense variants from the ClinVar database. Whole-cell patch-clamp recording was used to classify the function of 331 variants. Subsequently, we proposed 24 criteria for the pathogenicity interpretation of KCNQ2 variants and used them to assess pathogenicity of 331 variants. Finally, we analyzed the clinical phenotypes of patients carrying these variants, and explored the correlations between functional mechanisms and phenotypes. KEY FINDINGS In the homozygous state, 287 were classified as loss-of-function and 14 as gain-of-function. In the more clinically relative heterozygous state, 200 variants exhibited functional impairment, 121 of which showed dominant-negative effects on wild-type KCNQ2 subunits. After introducing functional data as strong-level evidence to interpret pathogenicity, over half of variants (169/331) were reclassified and 254 were classified as pathogenic/likely pathogenic. Moreover, dominant-negative effect and haploinsufficiency were identified as primary mechanisms in DEE/ID and SeLNE, respectively. The degree of impairment of channel function correlated with the phenotype severity. SIGNIFICANCE Our study reveals the possible cause of KCNQ2-related disorders at the molecular level, provides compelling evidence for clinical classification of KCNQ2 variants, and expands the knowledge of correlations between functional mechanisms and phenotypes.
Collapse
Affiliation(s)
- Yuwei Zhang
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuqing Xue
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yu Ma
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaonan Du
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Zhiqiang Yan
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
3
|
Borowicz-Reutt K, Czernia J, Krawczyk M. Genetic Background of Epilepsy and Antiepileptic Treatments. Int J Mol Sci 2023; 24:16280. [PMID: 38003469 PMCID: PMC10671416 DOI: 10.3390/ijms242216280] [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: 10/02/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Advanced identification of the gene mutations causing epilepsy syndromes is expected to translate into faster diagnosis and more effective treatment of these conditions. Over the last 5 years, approximately 40 clinical trials on the treatment of genetic epilepsies have been conducted. As a result, some medications that are not regular antiseizure drugs (e.g., soticlestat, fenfluramine, or ganaxolone) have been introduced to the treatment of drug-resistant seizures in Dravet, Lennox-Gastaut, maternally inherited chromosome 15q11.2-q13.1 duplication (Dup 15q) syndromes, and protocadherin 19 (PCDH 19)-clusterig epilepsy. And although the effects of soticlestat, fenfluramine, and ganaxolone are described as promising, they do not significantly affect the course of the mentioned epilepsy syndromes. Importantly, each of these syndromes is related to mutations in several genes. On the other hand, several mutations can occur within one gene, and different gene variants may be manifested in different disease phenotypes. This complex pattern of inheritance contributes to rather poor genotype-phenotype correlations. Hence, the detection of a specific mutation is not synonymous with a precise diagnosis of a specific syndrome. Bearing in mind that seizures develop as a consequence of the predominance of excitatory over inhibitory processes, it seems reasonable that mutations in genes encoding sodium and potassium channels, as well as glutamatergic and gamma-aminobutyric (GABA) receptors, play a role in the pathogenesis of epilepsy. In some cases, different pathogenic variants of the same gene can result in opposite functional effects, determining the effectiveness of therapy with certain medications. For instance, seizures related to gain-of-function (GoF) mutations in genes encoding sodium channels can be successfully treated with sodium channel blockers. On the contrary, the same drugs may aggravate seizures related to loss-of-function (LoF) variants of the same genes. Hence, knowledge of gene mutation-treatment response relationships facilitates more favorable selection of drugs for anticonvulsant therapy.
Collapse
Affiliation(s)
- Kinga Borowicz-Reutt
- Independent Unit of Experimental Neuropathophysiology, Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (J.C.); (M.K.)
| | | | | |
Collapse
|
4
|
Ye J, Tang S, Miao P, Gong Z, Shu Q, Feng J, Li Y. Clinical analysis and functional characterization of KCNQ2-related developmental and epileptic encephalopathy. Front Mol Neurosci 2023; 16:1205265. [PMID: 37497102 PMCID: PMC10366601 DOI: 10.3389/fnmol.2023.1205265] [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: 04/13/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Background Developmental and epileptic encephalopathy (DEE) is a condition characterized by severe seizures and a range of developmental impairments. Pathogenic variants in KCNQ2, encoding for potassium channel subunit, cause KCNQ2-related DEE. This study aimed to examine the relationships between genotype and phenotype in KCNQ2-related DEE. Methods In total, 12 patients were enrolled in this study for genetic testing, clinical analysis, and developmental evaluation. Pathogenic variants of KCNQ2 were characterized through a whole-cell electrophysiological recording expressed in Chinese hamster ovary (CHO) cells. The expression levels of the KCNQ2 subunit and its localization at the plasma membrane were determined using Western blot analysis. Results Seizures were detected in all patients. All DEE patients showed evidence of developmental delay. In total, 11 de novo KCNQ2 variants were identified, including 10 missense variants from DEE patients and one truncating variant from a patient with self-limited neonatal epilepsy (SeLNE). All variants were found to be loss of function through analysis of M-currents using patch-clamp recordings. The functional impact of variants on M-current in heteromericKCNQ2/3 channels may be associated with the severity of developmental disorders in DEE. The variants with dominant-negative effects in heteromeric channels may be responsible for the profound developmental phenotype. Conclusion The mechanism underlying KCNQ2-related DEE involves a reduction of the M-current through dominant-negative effects, and the severity of developmental disorders in DEE may be predicted by the impact of variants on the M-current of heteromericKCNQ2/3 channels.
Collapse
Affiliation(s)
- Jia Ye
- National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyang Tang
- Pediatric Department, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pu Miao
- Pediatric Department, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhefeng Gong
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Shu
- Pediatric Department, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Feng
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuezhou Li
- National Clinical Research Center for Child Health, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Pijpers JA, Au PYB, Weeke LC, Vein AA, Smit LS, Vilan A, Jacobs E, de Vries LS, Steggerda SJ, Cilio MR, Carapancea E, Cornet MC, Appendino JP, Peeters-Scholte CMPCD. Early recognition of characteristic conventional and amplitude-integrated EEG patterns of seizures in SCN2A and KCNQ3-related epilepsy in neonates. Seizure 2023; 110:212-219. [PMID: 37429183 DOI: 10.1016/j.seizure.2023.06.016] [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: 02/18/2023] [Revised: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 07/12/2023] Open
Abstract
PURPOSE Early recognition of seizures in neonates secondary to pathogenic variants in potassium or sodium channel coding genes is crucial, as these seizures are often resistant to commonly used anti-seizure medications but respond well to sodium channel blockers. Recently, a characteristic ictal amplitude-integrated electroencephalogram (aEEG) pattern was described in neonates with KCNQ2-related epilepsy. We report a similar aEEG pattern in seizures caused by SCN2A- and KCNQ3-pathogenic variants, as well as conventional EEG (cEEG) descriptions. METHODS International multicentre descriptive study, reporting clinical characteristics, aEEG and cEEG findings of 13 neonates with seizures due to pathogenic SCN2A- and KCNQ3-variants. As a comparison group, aEEGs and cEEGs of neonates with seizures due to hypoxic-ischemic encephalopathy (n = 117) and other confirmed genetic causes affecting channel function (n = 55) were reviewed. RESULTS In 12 out of 13 patients, the aEEG showed a characteristic sequence of brief onset with a decrease, followed by a quick rise, and then postictal amplitude attenuation. This pattern correlated with bilateral EEG onset attenuation, followed by rhythmic discharges ending in several seconds of post-ictal amplitude suppression. Apart from patients with KCNQ2-related epilepsy, none of the patients in the comparison groups had a similar aEEG or cEEG pattern. DISCUSSION Seizures in SCN2A- and KCNQ3-related epilepsy in neonates can usually be recognized by a characteristic ictal aEEG pattern, previously reported only in KCNQ2-related epilepsy, extending this unique feature to other channelopathies. Awareness of this pattern facilitates the prompt initiation of precision treatment with sodium channel blockers even before genetic results are available.
Collapse
Affiliation(s)
- Judith A Pijpers
- Departments of Neurology and Clinical Neurophysiology, Leiden University Medical Center, PO Box 9600, Leiden 2300 RC, the Netherlands.
| | - Ping Yee Billie Au
- Departments of Medical Genetics, Cumming School of Medicine, University of Calgary, Canada
| | - Lauren C Weeke
- Departments of Neonatology, Erasmus Medical Center, the Netherlands
| | - Alla A Vein
- Departments of Neurology and Clinical Neurophysiology, Leiden University Medical Center, PO Box 9600, Leiden 2300 RC, the Netherlands
| | - Liesbeth S Smit
- Departments of Neonatology, Erasmus Medical Center, the Netherlands; Departments of Neurology, Erasmus Medical Center, the Netherlands
| | - Ana Vilan
- Department of Neonatology, Centro Hospitalar Universitário São João, Faculty of Medicine, University of Porto, Portugal
| | - Elke Jacobs
- Departments of Neurology, Erasmus Medical Center, the Netherlands
| | - Linda S de Vries
- Departments of Neonatology, Leiden University Medical Center, the Netherlands
| | - Sylke J Steggerda
- Departments of Neonatology, Leiden University Medical Center, the Netherlands
| | - Maria Roberta Cilio
- Division of Pediatric Neurology, Department of Pediatrics, Catholic University of Louvain, Brussels, Belgium
| | - Evelina Carapancea
- Division of Pediatric Neurology, Department of Pediatrics, Catholic University of Louvain, Brussels, Belgium
| | - Marie-Coralie Cornet
- Division of Neonatology, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA, USA
| | - Juan P Appendino
- Departments of Pediatric Neurology, Cumming School of Medicine, University of Calgary, Canada
| | - Cacha M P C D Peeters-Scholte
- Departments of Neurology and Clinical Neurophysiology, Leiden University Medical Center, PO Box 9600, Leiden 2300 RC, the Netherlands
| |
Collapse
|
6
|
Sun S, Wang H. Clocking Epilepsies: A Chronomodulated Strategy-Based Therapy for Rhythmic Seizures. Int J Mol Sci 2023; 24:4223. [PMID: 36835631 PMCID: PMC9962262 DOI: 10.3390/ijms24044223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by hypersynchronous recurrent neuronal activities and seizures, as well as loss of muscular control and sometimes awareness. Clinically, seizures have been reported to display daily variations. Conversely, circadian misalignment and circadian clock gene variants contribute to epileptic pathogenesis. Elucidation of the genetic bases of epilepsy is of great importance because the genetic variability of the patients affects the efficacies of antiepileptic drugs (AEDs). For this narrative review, we compiled 661 epilepsy-related genes from the PHGKB and OMIM databases and classified them into 3 groups: driver genes, passenger genes, and undetermined genes. We discuss the potential roles of some epilepsy driver genes based on GO and KEGG analyses, the circadian rhythmicity of human and animal epilepsies, and the mutual effects between epilepsy and sleep. We review the advantages and challenges of rodents and zebrafish as animal models for epileptic studies. Finally, we posit chronomodulated strategy-based chronotherapy for rhythmic epilepsies, integrating several lines of investigation for unraveling circadian mechanisms underpinning epileptogenesis, chronopharmacokinetic and chronopharmacodynamic examinations of AEDs, as well as mathematical/computational modeling to help develop time-of-day-specific AED dosing schedules for rhythmic epilepsy patients.
Collapse
Affiliation(s)
- Sha Sun
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| |
Collapse
|
7
|
Li XT. The modulation of potassium channels by estrogens facilitates neuroprotection. Front Cell Dev Biol 2022; 10:998009. [PMID: 36393851 PMCID: PMC9643774 DOI: 10.3389/fcell.2022.998009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/11/2022] [Indexed: 08/31/2023] Open
Abstract
Estrogens, the sex hormones, have the potential to govern multiple cellular functions, such as proliferation, apoptosis, differentiation, and homeostasis, and to exert numerous beneficial influences for the cardiovascular system, nervous system, and bones in genomic and/or non-genomic ways. Converging evidence indicates that estrogens serve a crucial role in counteracting neurodegeneration and ischemic injury; they are thereby being considered as a potent neuroprotectant for preventing neurological diseases such as Alzheimer's disease and stroke. The underlying mechanism of neuroprotective effects conferred by estrogens is thought to be complex and multifactorial, and it remains obscure. It is well established that the K+ channels broadly expressed in a variety of neural subtypes determine the essential physiological features of neuronal excitability, and dysfunction of these channels is closely associated with diverse brain deficits, such as ataxia and epilepsy. A growing body of evidence supports a neuroprotective role of K+ channels in malfunctions of nervous tissues, with the channels even being a therapeutic target in clinical trials. As multitarget steroid hormones, estrogens also regulate the activity of distinct K+ channels to generate varying biological actions, and accumulated data delineate that some aspects of estrogen-mediated neuroprotection may arise from the impact on multiple K+ channels, including Kv, BK, KATP, and K2P channels. The response of these K+ channels after acute or chronic exposure to estrogens may oppose pathological abnormality in nervous cells, which serves to extend our understanding of these phenomena.
Collapse
Affiliation(s)
- Xian-Tao Li
- School of Medicine, Guizhou University, Guiyang, China
- Department of Neuroscience, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
8
|
Gao K, Lin Z, Wen S, Jiang Y. Potassium channels and epilepsy. Acta Neurol Scand 2022; 146:699-707. [PMID: 36225112 DOI: 10.1111/ane.13695] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 01/10/2023]
Abstract
With the development and application of next-generation sequencing technology, the aetiological diagnosis of genetic epilepsy is rapidly becoming easier and less expensive. Additionally, there is a growing body of research into precision therapy based on genetic diagnosis. The numerous genes in the potassium ion channel family constitute the largest family of ion channels: this family is divided into different subtypes. Potassium ion channels play a crucial role in the electrical activity of neurons and are directly involved in the mechanism of epileptic seizures. In China, scientific research on genetic diagnosis and studies of precision therapy for genetic epilepsy are progressing rapidly. Many cases of epilepsy caused by mutation of potassium channel genes have been identified, and several potassium channel gene targets and drug candidates have been discovered. The purpose of this review is to briefly summarize the progress of research on the precise diagnosis and treatment of potassium ion channel-related genetic epilepsy, especially the research conducted in China. Here in, we review several large cohort studies on the genetic diagnosis of epilepsy in China in recent years, summarized the proportion of potassium channel genes. We focus on the progress of precison therapy on some hot epilepsy related potassium channel genes: KCNA1, KCNA2, KCNB1, KCNC1, KCND2, KCNQ2, KCNQ3, KCNMA1, and KCNT1.
Collapse
Affiliation(s)
- Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.,Children Epilepsy Center, Peking University First Hospital, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Zehong Lin
- Department of Neurology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Sijia Wen
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.,Children Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.,Children Epilepsy Center, Peking University First Hospital, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
9
|
Edmond MA, Hinojo-Perez A, Wu X, Perez Rodriguez ME, Barro-Soria R. Distinctive mechanisms of epilepsy-causing mutants discovered by measuring S4 movement in KCNQ2 channels. eLife 2022; 11:77030. [PMID: 35642783 PMCID: PMC9197397 DOI: 10.7554/elife.77030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/31/2022] [Indexed: 01/10/2023] Open
Abstract
Neuronal KCNQ channels mediate the M-current, a key regulator of membrane excitability in the central and peripheral nervous systems. Mutations in KCNQ2 channels cause severe neurodevelopmental disorders, including epileptic encephalopathies. However, the impact that different mutations have on channel function remains poorly defined, largely because of our limited understanding of the voltage-sensing mechanisms that trigger channel gating. Here, we define the parameters of voltage sensor movements in wt-KCNQ2 and channels bearing epilepsy-associated mutations using cysteine accessibility and voltage clamp fluorometry (VCF). Cysteine modification reveals that a stretch of eight to nine amino acids in the S4 becomes exposed upon voltage sensing domain activation of KCNQ2 channels. VCF shows that the voltage dependence and the time course of S4 movement and channel opening/closing closely correlate. VCF reveals different mechanisms by which different epilepsy-associated mutations affect KCNQ2 channel voltage-dependent gating. This study provides insight into KCNQ2 channel function, which will aid in uncovering the mechanisms underlying channelopathies.
Collapse
Affiliation(s)
- Michaela A Edmond
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, United States
| | - Andy Hinojo-Perez
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, United States
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, United States
| | - Marta E Perez Rodriguez
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, United States
| | - Rene Barro-Soria
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, United States
| |
Collapse
|
10
|
Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int J Mol Sci 2022; 23:ijms23084413. [PMID: 35457230 PMCID: PMC9028019 DOI: 10.3390/ijms23084413] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3β is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3β is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3β acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3β. Here, we discuss the direct and indirect mechanisms by which GSK3β phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3β activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3β-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.
Collapse
|
11
|
Zhang HXB, Heckman L, Niday Z, Jo S, Fujita A, Shim J, Pandey R, Al Jandal H, Jayakar S, Barrett LB, Smith J, Woolf CJ, Bean BP. Cannabidiol activates neuronal Kv7 channels. eLife 2022; 11:73246. [PMID: 35179483 PMCID: PMC8856652 DOI: 10.7554/elife.73246] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 02/09/2022] [Indexed: 01/10/2023] Open
Abstract
Cannabidiol (CBD), a chemical found in the Cannabis sativa plant, is a clinically effective antiepileptic drug whose mechanism of action is unknown. Using a fluorescence-based thallium flux assay, we performed a large-scale screen and found enhancement of flux through heterologously expressed human Kv7.2/7.3 channels by CBD. Patch-clamp recordings showed that CBD acts at submicromolar concentrations to shift the voltage dependence of Kv7.2/7.3 channels in the hyperpolarizing direction, producing a dramatic enhancement of current at voltages near –50 mV. CBD enhanced native M-current in mouse superior cervical ganglion starting at concentrations of 30 nM and also enhanced M-current in rat hippocampal neurons. The potent enhancement of Kv2/7.3 channels by CBD may contribute to its effectiveness as an antiepileptic drug by reducing neuronal hyperexcitability.
Collapse
Affiliation(s)
| | - Laurel Heckman
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Zachary Niday
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Akie Fujita
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jaehoon Shim
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Roshan Pandey
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Hoor Al Jandal
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Selwyn Jayakar
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Lee B Barrett
- F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Jennifer Smith
- ICCB-Longwood Screening Facility and Department of Immunology, Harvard Medical School, Boston, United States
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, United States.,F.M. Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
12
|
Ziobro JM, Eschbach K, Shellhaas RA. Novel Therapeutics for Neonatal Seizures. Neurotherapeutics 2021; 18:1564-1581. [PMID: 34386906 PMCID: PMC8608938 DOI: 10.1007/s13311-021-01085-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/04/2023] Open
Abstract
Neonatal seizures are a common neurologic emergency for which therapies have not significantly changed in decades. Improvements in diagnosis and pathophysiologic understanding of the distinct features of acute symptomatic seizures and neonatal-onset epilepsies present exceptional opportunities for development of precision therapies with potential to improve outcomes. Herein, we discuss the pathophysiology of neonatal seizures and review the evidence for currently available treatment. We present emerging therapies in clinical and preclinical development for the treatment of acute symptomatic neonatal seizures. Lastly, we discuss the role of precision therapies for genetic neonatal-onset epilepsies and address barriers and goals for developing new therapies for clinical care.
Collapse
Affiliation(s)
- Julie M Ziobro
- Department of Pediatrics, Michigan Medicine, C.S. Mott Children's Hospital, University of Michigan, 1540 E. Hospital Dr, Ann Arbor, MI, USA.
| | - Krista Eschbach
- Department of Pediatrics, Section of Neurology, Denver Anschutz School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, CO, 80045, USA
| | - Renée A Shellhaas
- Department of Pediatrics, Michigan Medicine, C.S. Mott Children's Hospital, University of Michigan, 1540 E. Hospital Dr, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Urrutia J, Aguado A, Gomis-Perez C, Muguruza-Montero A, Ballesteros OR, Zhang J, Nuñez E, Malo C, Chung HJ, Leonardo A, Bergara A, Villarroel A. An epilepsy-causing mutation leads to co-translational misfolding of the Kv7.2 channel. BMC Biol 2021; 19:109. [PMID: 34020651 PMCID: PMC8138981 DOI: 10.1186/s12915-021-01040-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 04/29/2021] [Indexed: 12/21/2022] Open
Abstract
Background The amino acid sequence of proteins generally carries all the necessary information for acquisition of native conformations, but the vectorial nature of translation can additionally determine the folding outcome. Such consideration is particularly relevant in human diseases associated to inherited mutations leading to structural instability, aggregation, and degradation. Mutations in the KCNQ2 gene associated with human epilepsy have been suggested to cause misfolding of the encoded Kv7.2 channel. Although the effect on folding of mutations in some domains has been studied, little is known of the way pathogenic variants located in the calcium responsive domain (CRD) affect folding. Here, we explore how a Kv7.2 mutation (W344R) located in helix A of the CRD and associated with hereditary epilepsy interferes with channel function. Results We report that the epilepsy W344R mutation within the IQ motif of CRD decreases channel function, but contrary to other mutations at this site, it does not impair the interaction with Calmodulin (CaM) in vitro, as monitored by multiple in vitro binding assays. We find negligible impact of the mutation on the structure of the complex by molecular dynamic computations. In silico studies revealed two orientations of the side chain, which are differentially populated by WT and W344R variants. Binding to CaM is impaired when the mutated protein is produced in cellulo but not in vitro, suggesting that this mutation impedes proper folding during translation within the cell by forcing the nascent chain to follow a folding route that leads to a non-native configuration, and thereby generating non-functional ion channels that fail to traffic to proper neuronal compartments. Conclusions Our data suggest that the key pathogenic mechanism of Kv7.2 W344R mutation involves the failure to adopt a configuration that can be recognized by CaM in vivo but not in vitro. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01040-1.
Collapse
Affiliation(s)
- Janire Urrutia
- Instituto Biofisika, CSIC-UPV/EHU, 48940, Leioa, Spain.,Present address: Department of Physiology, Faculty of Medicine and Nursery, UPV/EHU, 48940, Leioa, Spain
| | | | - Carolina Gomis-Perez
- Instituto Biofisika, CSIC-UPV/EHU, 48940, Leioa, Spain.,Present address: Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Jiaren Zhang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Eider Nuñez
- Instituto Biofisika, CSIC-UPV/EHU, 48940, Leioa, Spain
| | | | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aritz Leonardo
- Departamento de Física Aplicada II, Universidad del País Vasco, UPV/EHU, 48940, Leioa, Spain.,Donostia International Physics Center, 20018, Donostia, Spain
| | - Aitor Bergara
- Centro de Física de Materiales CFM, CSIC-UPV/EHU, 20018, Donostia, Spain.,Donostia International Physics Center, 20018, Donostia, Spain.,Departmento de Materia Condensada, Universidad del País Vasco, UPV/EHU, 48940, Leioa, Spain
| | | |
Collapse
|
14
|
Khayachi A, Schorova L, Alda M, Rouleau GA, Milnerwood AJ. Posttranslational modifications & lithium's therapeutic effect-Potential biomarkers for clinical responses in psychiatric & neurodegenerative disorders. Neurosci Biobehav Rev 2021; 127:424-445. [PMID: 33971223 DOI: 10.1016/j.neubiorev.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/14/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
Several neurodegenerative diseases and neuropsychiatric disorders display aberrant posttranslational modifications (PTMs) of one, or many, proteins. Lithium treatment has been used for mood stabilization for many decades, and is highly effective for large subsets of patients with diverse neurological conditions. However, the differential effectiveness and mode of action are not fully understood. In recent years, studies have shown that lithium alters several protein PTMs, altering their function, and consequently neuronal physiology. The impetus for this review is to outline the links between lithium's therapeutic mode of action and PTM homeostasis. We first provide an overview of the principal PTMs affected by lithium. We then describe several neuropsychiatric disorders in which PTMs have been implicated as pathogenic. For each of these conditions, we discuss lithium's clinical use and explore the putative mechanism of how it restores PTM homeostasis, and thereby cellular physiology. Evidence suggests that determining specific PTM patterns could be a promising strategy to develop biomarkers for disease and lithium responsiveness.
Collapse
Affiliation(s)
- A Khayachi
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| | - L Schorova
- McGill University Health Center Research Institute, Montréal, Quebec, Canada
| | - M Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - G A Rouleau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada; Department of Human Genetics, McGill University, Montréal, Quebec, Canada.
| | - A J Milnerwood
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
15
|
Ritter DM, Horn PS, Holland KD. In Silico Predictions of KCNQ Variant Pathogenicity in Epilepsy. Pediatr Neurol 2021; 118:48-54. [PMID: 33784504 PMCID: PMC8076079 DOI: 10.1016/j.pediatrneurol.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Variants in KCNQ2 and KCNQ3 may cause benign neonatal familial seizures and early infantile epileptic encephalopathy. Previous reports suggest that in silico models cannot predict pathogenicity accurately enough for clinical use. Here we sought to establish a model to accurately predict the pathogenicity of KCNQ2 and KCNQ3 missense variants based on available in silico prediction models. METHODS ClinVar and gnomAD databases of reported KCNQ2 and KCNQ3 missense variants in patients with neonatal epilepsy were accessed and classified as benign, pathogenic, or of uncertain significance. Sensitivity, specificity, and classification accuracy for prediction of pathogenicity were determined and compared for 10 widely used prediction algorithms program. A mathematical model of the variants (KCNQ Index) was created using their amino acid location and prediction algorithm scores to improve prediction accuracy. RESULTS Using clinically characterized variants, the free online tool PROVEAN accurately predicted pathogenicity 92% of the time and the KCNQ Index had an accuracy of 96%. However, when including the gnomAD database as benign variants, only the KCNQ Index was able to predict pathogenicity with an accuracy greater than 90% (sensitivity = 93% and specificity = 98%). No model could accurately predict the phenotype of variants. CONCLUSION We show that KCNQ channel variant pathogenicity can be predicted by a novel KCNQ Index in neonatal epilepsy. However, more work is needed to accurately predict the patient's epilepsy phenotype from in silico algorithms.
Collapse
Affiliation(s)
- David M Ritter
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Paul S Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Katherine D Holland
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
16
|
Borgini M, Mondal P, Liu R, Wipf P. Chemical modulation of Kv7 potassium channels. RSC Med Chem 2021; 12:483-537. [PMID: 34046626 PMCID: PMC8128042 DOI: 10.1039/d0md00328j] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/01/2020] [Indexed: 01/10/2023] Open
Abstract
The rising interest in Kv7 modulators originates from their ability to evoke fundamental electrophysiological perturbations in a tissue-specific manner. A large number of therapeutic applications are, in part, based on the clinical experience with two broad-spectrum Kv7 agonists, flupirtine and retigabine. Since precise molecular structures of human Kv7 channel subtypes in closed and open states have only very recently started to emerge, computational studies have traditionally been used to analyze binding modes and direct the development of more potent and selective Kv7 modulators with improved safety profiles. Herein, the synthetic and medicinal chemistry of small molecule modulators and the representative biological properties are summarized. Furthermore, new therapeutic applications supported by in vitro and in vivo assay data are suggested.
Collapse
Affiliation(s)
- Matteo Borgini
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Pravat Mondal
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Ruiting Liu
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| |
Collapse
|
17
|
Dirkx N, Miceli F, Taglialatela M, Weckhuysen S. The Role of Kv7.2 in Neurodevelopment: Insights and Gaps in Our Understanding. Front Physiol 2020; 11:570588. [PMID: 33192566 PMCID: PMC7657400 DOI: 10.3389/fphys.2020.570588] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Abstract
Kv7.2 subunits encoded by the KCNQ2 gene constitute a critical molecular component of the M-current, a subthreshold voltage-gated potassium current controlling neuronal excitability by dampening repetitive action potential firing. Pathogenic loss-of-function variants in KCNQ2 have been linked to epilepsy since 1998, and there is ample functional evidence showing that dysfunction of the channel indeed results in neuronal hyperexcitability. The recent description of individuals with severe developmental delay with or without seizures due to pathogenic variants in KCNQ2 (KCNQ2-encephalopathy) reveals that Kv7.2 channels also have an important role in neurodevelopment. Kv7.2 channels are expressed already very early in the developing brain when key developmental processes such as proliferation, differentiation, and synaptogenesis play a crucial role in brain morphogenesis and maturation. In this review, we will discuss the available evidence for a role of Kv7.2 channels in these neurodevelopmental processes, focusing in particular on insights derived from KCNQ2-related human phenotypes, from the spatio-temporal expression of Kv7.2 and other Kv7 family member, and from cellular and rodent models, highlighting critical gaps and research strategies to be implemented in the future. Lastly, we propose a model which divides the M-current activity in three different developmental stages, correlating with the cell characteristics during these particular periods in neuronal development, and how this can be linked with KCNQ2-related disorders. Understanding these mechanisms can create opportunities for new targeted therapies for KCNQ2-encephalopathy.
Collapse
Affiliation(s)
- Nina Dirkx
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, Antwerp, Belgium
| | - Francesco Miceli
- Section of Pharmacology, Department of Neuroscience, University of Naples Federico II, Naples, Italy
| | - Maurizio Taglialatela
- Section of Pharmacology, Department of Neuroscience, University of Naples Federico II, Naples, Italy
| | - Sarah Weckhuysen
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, Antwerp, Belgium.,Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
18
|
Li J, Maghera J, Lamothe SM, Marco EJ, Kurata HT. Heteromeric Assembly of Truncated Neuronal Kv7 Channels: Implications for Neurologic Disease and Pharmacotherapy. Mol Pharmacol 2020; 98:192-202. [DOI: 10.1124/mol.120.119644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
|
19
|
Gawel K, Langlois M, Martins T, van der Ent W, Tiraboschi E, Jacmin M, Crawford AD, Esguerra CV. Seizing the moment: Zebrafish epilepsy models. Neurosci Biobehav Rev 2020; 116:1-20. [PMID: 32544542 DOI: 10.1016/j.neubiorev.2020.06.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Zebrafish are now widely accepted as a valuable animal model for a number of different central nervous system (CNS) diseases. They are suitable both for elucidating the origin of these disorders and the sequence of events culminating in their onset, and for use as a high-throughput in vivo drug screening platform. The availability of powerful and effective techniques for genome manipulation allows the rapid modelling of different genetic epilepsies and of conditions with seizures as a core symptom. With this review, we seek to summarize the current knowledge about existing epilepsy/seizures models in zebrafish (both pharmacological and genetic) and compare them with equivalent rodent and human studies. New findings obtained from the zebrafish models are highlighted. We believe that this comprehensive review will highlight the value of zebrafish as a model for investigating different aspects of epilepsy and will help researchers to use these models to their full extent.
Collapse
Affiliation(s)
- Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland
| | | | - Teresa Martins
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Wietske van der Ent
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Ettore Tiraboschi
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Neurophysics Group, Center for Mind/Brain Sciences, University of Trento, Piazza Manifattura 1, Building 14, 38068, Rovereto, TN, Italy
| | - Maxime Jacmin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg; Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway.
| |
Collapse
|
20
|
Zhao L, Luo F, Wang A, Zhang J, Wang Y, Zhao L, Wang Z, Pu Q. Quick stabilization of capillary for rapid determination of potassium ions in the blood of epilepsy patients by capillary electrophoresis without sample pretreatment. Electrophoresis 2020; 41:1273-1279. [PMID: 32358896 DOI: 10.1002/elps.202000022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 11/07/2022]
Abstract
Mutations in the potassium channel genes may be linked to the development of epilepsy and affect the blood potassium levels. Therefore, accurate determination of potassium in the blood will be critical to diagnose the cause of epilepsy. CE is a competent technique for the fast detection of multiple ions, but complicated matrices of a blood sample may cause significant variation of migration times and the peak shape. In this work, a procedure for rapid stabilization of the capillary inner surface through preflushing of a blood sample was employed. The process takes only 40 min for a capillary and then it can be used for more than 2 weeks. No pretreatment of the blood sample or other surface modification of the capillary is needed for the analysis. The RSDs of the migration time and peak area were reduced to 1.5 and 5.1% from 12.6 and 14.5%, respectively. The proposed method has been successfully applied to the determination of the potassium contents in the blood sample of patients with epilepsy at different stages. The recoveries of potassium ions in these blood samples are in a range from 86.5 to 104.5%.
Collapse
Affiliation(s)
- Litao Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, P. R. China
| | - Fanghong Luo
- School of Pharmacy, Lanzhou University, Lanzhou, P. R. China
| | - Anting Wang
- School of Pharmacy, Lanzhou University, Lanzhou, P. R. China
| | - Jing Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, P. R. China
| | - Yuanhang Wang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, P. R. China
| | - Liangtao Zhao
- TSing Biomedical Research Center, The Second Hospital of Lanzhou University, Lanzhou, P. R. China
| | - Zhaoyan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, P. R. China
| | - Qiaosheng Pu
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, P. R. China
| |
Collapse
|
21
|
Hedrich UBS, Lauxmann S, Lerche H. SCN2A channelopathies: Mechanisms and models. Epilepsia 2020; 60 Suppl 3:S68-S76. [PMID: 31904120 DOI: 10.1111/epi.14731] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/21/2019] [Indexed: 01/01/2023]
Abstract
Variants in the SCN2A gene, encoding the voltage-gated sodium channel NaV 1.2, cause a variety of neuropsychiatric syndromes with different severity ranging from self-limiting epilepsies with early onset to developmental and epileptic encephalopathy with early or late onset and intellectual disability (ID), as well as ID or autism without seizures. Functional analysis of channel defects demonstrated a genotype-phenotype correlation and suggested effective treatment options for one group of affected patients carrying gain-of-function variants. Here, we sum up the functional mechanisms underlying different phenotypes of patients with SCN2A channelopathies and present currently available models that can help in understanding SCN2A-related disorders.
Collapse
Affiliation(s)
- Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
Zhang J, Kim EC, Chen C, Procko E, Pant S, Lam K, Patel J, Choi R, Hong M, Joshi D, Bolton E, Tajkhorshid E, Chung HJ. Identifying mutation hotspots reveals pathogenetic mechanisms of KCNQ2 epileptic encephalopathy. Sci Rep 2020; 10:4756. [PMID: 32179837 PMCID: PMC7075958 DOI: 10.1038/s41598-020-61697-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/02/2020] [Indexed: 11/08/2022] Open
Abstract
Kv7 channels are enriched at the axonal plasma membrane where their voltage-dependent potassium currents suppress neuronal excitability. Mutations in Kv7.2 and Kv7.3 subunits cause epileptic encephalopathy (EE), yet the underlying pathogenetic mechanism is unclear. Here, we used novel statistical algorithms and structural modeling to identify EE mutation hotspots in key functional domains of Kv7.2 including voltage sensing S4, the pore loop and S6 in the pore domain, and intracellular calmodulin-binding helix B and helix B-C linker. Characterization of selected EE mutations from these hotspots revealed that L203P at S4 induces a large depolarizing shift in voltage dependence of Kv7.2 channels and L268F at the pore decreases their current densities. While L268F severely reduces expression of heteromeric channels in hippocampal neurons without affecting internalization, K552T and R553L mutations at distal helix B decrease calmodulin-binding and axonal enrichment. Importantly, L268F, K552T, and R553L mutations disrupt current potentiation by increasing phosphatidylinositol 4,5-bisphosphate (PIP2), and our molecular dynamics simulation suggests PIP2 interaction with these residues. Together, these findings demonstrate that each EE variant causes a unique combination of defects in Kv7 channel function and neuronal expression, and suggest a critical need for both prediction algorithms and experimental interrogations to understand pathophysiology of Kv7-associated EE.
Collapse
Affiliation(s)
- Jiaren Zhang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Eung Chang Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Congcong Chen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Erik Procko
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Shashank Pant
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Kin Lam
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Jaimin Patel
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Rebecca Choi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Mary Hong
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Dhruv Joshi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Eric Bolton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Emad Tajkhorshid
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA.
| |
Collapse
|
23
|
Sills GJ, Rogawski MA. Mechanisms of action of currently used antiseizure drugs. Neuropharmacology 2020; 168:107966. [PMID: 32120063 DOI: 10.1016/j.neuropharm.2020.107966] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 02/08/2023]
Abstract
Antiseizure drugs (ASDs) prevent the occurrence of seizures; there is no evidence that they have disease-modifying properties. In the more than 160 years that orally administered ASDs have been available for epilepsy therapy, most agents entering clinical practice were either discovered serendipitously or with the use of animal seizure models. The ASDs originating from these approaches act on brain excitability mechanisms to interfere with the generation and spread of epileptic hyperexcitability, but they do not address the specific defects that are pathogenic in the epilepsies for which they are prescribed, which in most cases are not well understood. There are four broad classes of such ASD mechanisms: (1) modulation of voltage-gated sodium channels (e.g. phenytoin, carbamazepine, lamotrigine), voltage-gated calcium channels (e.g. ethosuximide), and voltage-gated potassium channels [e.g. retigabine (ezogabine)]; (2) enhancement of GABA-mediated inhibitory neurotransmission (e.g. benzodiazepines, tiagabine, vigabatrin); (3) attenuation of glutamate-mediated excitatory neurotransmission (e.g. perampanel); and (4) modulation of neurotransmitter release via a presynaptic action (e.g. levetiracetam, brivaracetam, gabapentin, pregabalin). In the past two decades there has been great progress in identifying the pathophysiological mechanisms of many genetic epilepsies. Given this new understanding, attempts are being made to engineer specific small molecule, antisense and gene therapies that functionally reverse or structurally correct pathogenic defects in epilepsy syndromes. In the near future, these new therapies will begin a paradigm shift in the treatment of some rare genetic epilepsy syndromes, but targeted therapies will remain elusive for the vast majority of epilepsies until their causes are identified. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Graeme J Sills
- School of Life Sciences, University of Glasgow, Glasgow, UK.
| | - Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Department of Pharmacology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
24
|
Li S, Sunchen S, He D, Qin C, Zuo Z, Shen B, Cao Z, Hong W, Miao L. ImKTx96, a peptide blocker of the Kv1.2 ion channel from the venom of the scorpion Isometrus maculates. Peptides 2020; 123:170172. [PMID: 31626826 DOI: 10.1016/j.peptides.2019.170172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/13/2023]
Abstract
Scorpion venom contains diverse bioactive peptides that can recognize and interact with membrane proteins such as ion channels. These natural toxins are believed to be useful tools for exploring the structure and function of ion channels. In this study, we characterized a K+-channel toxin gene, ImKTx96, from the venom gland cDNA library of the scorpion Isometrus maculates. The peptide deduced from the ImKTx96 precursor nucleotide sequence contains a signal peptide of 27 amino acid residues and a mature peptide of 29 residues with three disulfide bridges. Multiple sequence alignment indicated that ImKTx96 is similar with the scorpion toxins that typically target K+-channels. The recombined ImKTx96 peptide (rImKTx96) was expressed in the Escherichia coli system, and purified by GST-affinity chromatography and RP-HPLC. Results from whole-cell patch-clamp experiments revealed that rImKTx96 can inhibit the current of the Kv1.2 ion channel expressed in HEK293 cells. The 3D structure of ImKTx96 was constructed by molecular modeling, and the complex formed by ImKTx96 interacting with the Kv1.2 ion channel was obtained by molecular docking. Based on its structural features and pharmacological functions, ImKTx96 was identified as one member of K+-channel scorpion toxin α-KTx10 group and may be useful as a molecular probe for investigating the structure and function of the Kv1.2 ion channel.
Collapse
Affiliation(s)
- Sipian Li
- Department of Biochemistry, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Shuwen Sunchen
- Department of Biochemistry, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Dangui He
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China
| | - Chenhu Qin
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China
| | - Zheng Zuo
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China
| | - Bingzheng Shen
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China
| | - Zhijian Cao
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China; Hubei Province Engineering and Technology Research, Center for Fluorinated Pharmaceuticals, Wuhan University, Wuhan 430072, PR China
| | - Wei Hong
- State Key Laboratory of Virology, Renmin Hospital, Wuhan University, Wuhan 430072, PR China.
| | - Lixia Miao
- Department of Biochemistry, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
25
|
Gan J, Cai Q, Galer P, Ma D, Chen X, Huang J, Bao S, Luo R. Mapping the knowledge structure and trends of epilepsy genetics over the past decade: A co-word analysis based on medical subject headings terms. Medicine (Baltimore) 2019; 98:e16782. [PMID: 31393404 PMCID: PMC6709143 DOI: 10.1097/md.0000000000016782] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Over the past 10 years, epilepsy genetics has made dramatic progress. This study aimed to analyze the knowledge structure and the advancement of epilepsy genetics over the past decade based on co-word analysis of medical subject headings (MeSH) terms. METHODS Scientific publications focusing on epilepsy genetics from the PubMed database (January 2009-December 2018) were retrieved. Bibliometric information was analyzed quantitatively using Bibliographic Item Co-Occurrence Matrix Builder (BICOMB) software. A knowledge social network analysis and publication trend based on the high-frequency MeSH terms was built using VOSviewer. RESULTS According to the search strategy, a total of 5185 papers were included. Among all the extracted MeSH terms, 86 high-frequency MeSH terms were identified. Hot spots were clustered into 5 categories including: "ion channel diseases," "beyond ion channel diseases," "experimental research & epigenetics," "single nucleotide polymorphism & pharmacogenetics," and "genetic techniques". "Epilepsy," "mutation," and "seizures," were located at the center of the knowledge network. "Ion channel diseases" are typically in the most prominent position of epilepsy genetics research. "Beyond ion channel diseases" and "genetic techniques," however, have gradually grown into research cores and trends, such as "intellectual disability," "infantile spasms," "phenotype," "exome," " deoxyribonucleic acid (DNA) copy number variations," and "application of next-generation sequencing." While ion channel genes such as "SCN1A," "KCNQ2," "SCN2A," "SCN8A" accounted for nearly half of epilepsy genes in MeSH terms, a number of additional beyond ion channel genes like "CDKL5," "STXBP1," "PCDH19," "PRRT2," "LGI1," "ALDH7A1," "MECP2," "EPM2A," "ARX," "SLC2A1," and more were becoming increasingly popular. In contrast, gene therapies, treatment outcome, and genotype-phenotype correlations were still in their early stages of research. CONCLUSION This co-word analysis provides an overview of epilepsy genetics research over the past decade. The 5 research categories display publication hot spots and trends in epilepsy genetics research which could consequently supply some direction for geneticists and epileptologists when launching new projects.
Collapse
Affiliation(s)
- Jing Gan
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, China
| | - Qianyun Cai
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, China
| | - Peter Galer
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, PA
| | - Dan Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
| | - Xiaolu Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
| | - Jichong Huang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, China
| | - Shan Bao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, China
| | - Rong Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu
| |
Collapse
|
26
|
Yu X, Yang L, Li J, Li W, Li D, Wang R, Wu K, Chen W, Zhang Y, Qiu Z, Zhou W. De Novo and Inherited SETD1A Variants in Early-onset Epilepsy. Neurosci Bull 2019; 35:1045-1057. [PMID: 31197650 DOI: 10.1007/s12264-019-00400-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022] Open
Abstract
Early-onset epilepsy is a neurological abnormality in childhood, and it is especially common in the first 2 years after birth. Seizures in early life mostly result from structural or metabolic disorders in the brain, and the genetic causes of idiopathic seizures have been extensively investigated. In this study, we identified four missense mutations in the SETD1A gene (SET domain-containing 1A, histone lysine methyltransferase): three de novo mutations in three individuals and one inherited mutation in a four-generation family. Whole-exome sequencing indicated that all four of these mutations were responsible for the seizures. Mutations of SETD1A have been implicated in schizophrenia and developmental disorders, so we examined the role of the four mutations (R913C, Q269R, G1369R, and R1392H) in neural development. We found that their expression in mouse primary cortical neurons affected excitatory synapse development. Moreover, expression of the R913C mutation also affected the migration of cortical neurons in the mouse brain. We further identified two common genes (Neurl4 and Usp39) affected by mutations of SETD1A. These results suggested that the mutations of SETD1A play a fundamental role in abnormal synaptic function and the development of neurons, so they may be pathogenic factors for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiuya Yu
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Lin Yang
- Clinical Genetic Center, Children's Hospital of Fudan University, Shanghai, 201102, China.,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jin Li
- Institute of Biomedicine Sciences, Fudan University, Shanghai, 200032, China
| | - Wanxing Li
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Dongzhi Li
- Department of Prenatal Diagnosis, The Women and Children's Medical Center, Guangzhou, 510623, China
| | - Ran Wang
- Euler Genomics, Beijing, 102206, China
| | - Kai Wu
- Euler Genomics, Beijing, 102206, China
| | | | - Yi Zhang
- Euler Genomics, Beijing, 102206, China. .,School of Life Sciences, Peking University, Beijing, 100191, China.
| | - Zilong Qiu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Wenhao Zhou
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
27
|
Lin Z, Huang X, Zhou W, Zhang W, Liu Y, Bian T, Niu L, Meng L, Guo Y. Ultrasound Stimulation Modulates Voltage-Gated Potassium Currents Associated With Action Potential Shape in Hippocampal CA1 Pyramidal Neurons. Front Pharmacol 2019; 10:544. [PMID: 31178727 PMCID: PMC6538798 DOI: 10.3389/fphar.2019.00544] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/30/2019] [Indexed: 11/28/2022] Open
Abstract
Potassium channels (K+) play an important role in the regulation of cellular signaling. Dysfunction of potassium channels is associated with several severe ion channels diseases, such as long QT syndrome, episodic ataxia and epilepsy. Ultrasound stimulation has proven to be an effective non-invasive tool for the modulation of ion channels and neural activity. In this study, we demonstrate that ultrasound stimulation enables to modulate the potassium currents and has an impact on the shape modulation of action potentials (AP) in the hippocampal pyramidal neurons using whole-cell patch-clamp recordings in vitro. The results show that outward potassium currents in neurons increase significantly, approximately 13%, in response to 30 s ultrasound stimulation. Simultaneously, the increasing outward potassium currents directly decrease the resting membrane potential (RMP) from −64.67 ± 1.10 mV to −67.51 ± 1.35 mV. Moreover, the threshold current and AP fall rate increase while the reduction of AP half-width and after-hyperpolarization peak time is detected. During ultrasound stimulation, reduction of the membrane input resistance of pyramidal neurons can be found and shorter membrane time constant is achieved. Additionally, we verify that the regulation of potassium currents and shape of action potential is mainly due to the mechanical effects induced by ultrasound. Therefore, ultrasound stimulation may offer an alternative tool to treat some ion channels diseases related to potassium channels.
Collapse
Affiliation(s)
- Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaowei Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wenjun Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of E&M, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Yingzhe Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Tianyuan Bian
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Knauer B, Yoshida M. Switching between persistent firing and depolarization block in individual rat CA1 pyramidal neurons. Hippocampus 2019; 29:817-835. [PMID: 30794330 DOI: 10.1002/hipo.23078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/22/2018] [Accepted: 01/15/2019] [Indexed: 11/07/2022]
Abstract
The hippocampal formation plays a role in mnemonic tasks and epileptic discharges in vivo. In vitro, these functions and malfunctions may relate to persistent firing (PF) and depolarization block (DB), respectively. Pyramidal neurons of the CA1 field have previously been reported to engage in either PF or DB during cholinergic stimulation. However, it is unknown whether these cells constitute disparate populations of neurons. Furthermore, it is unclear which cell-specific peculiarities may mediate their diverse response properties. However, it has not been shown whether individual CA1 pyramidal neurons can switch between PF and DB states. Here, we used whole cell patch clamp in the current clamp mode on in vitro CA1 pyramidal neurons from acutely sliced rat tissue to test various intrinsic properties which may provoke individual cells to switch between PF and DB. We found that individual cells could switch from PF to DB, in a cholinergic agonist concentration dependent manner and depending on the parameters of stimulation. We also demonstrate involvement of TRPC and potassium channels in this switching. Finally, we report that the probability for DB was more pronounced in the proximal than in the distal half of CA1. These findings offer a potential mechanism for the stronger spatial modulation in proximal, compared to distal CA1, as place field formation was shown to be affected by DB. Taken together, our results suggest that PF and DB are not mutually exclusive response properties of individual neurons. Rather, a cell's response mode depends on a variety of intrinsic properties, and modulation of these properties enables switching between PF and DB.
Collapse
Affiliation(s)
- Beate Knauer
- International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
- Faculty of Psychology, Mercator Research Group - Structure of Memory, Ruhr University Bochum, Bochum, Germany
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Motoharu Yoshida
- Faculty of Psychology, Mercator Research Group - Structure of Memory, Ruhr University Bochum, Bochum, Germany
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
29
|
Barro-Soria R. Epilepsy-associated mutations in the voltage sensor of KCNQ3 affect voltage dependence of channel opening. J Gen Physiol 2018; 151:247-257. [PMID: 30578330 PMCID: PMC6363412 DOI: 10.1085/jgp.201812221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/03/2018] [Indexed: 11/20/2022] Open
Abstract
One of the major factors known to cause neuronal hyperexcitability is malfunction of the potassium channels formed by KCNQ2 and KCNQ3. These channel subunits underlie the M current, which regulates neuronal excitability. Here, I investigate the molecular mechanisms by which epilepsy-associated mutations in the voltage sensor (S4) of KCNQ3 cause channel malfunction. Voltage clamp fluorometry reveals that the R230C mutation in KCNQ3 allows S4 movement but shifts the open/closed transition of the gate to very negative potentials. This results in the mutated channel remaining open throughout the physiological voltage range. Substitution of R230 with natural and unnatural amino acids indicates that the functional effect of the arginine residue at position 230 depends on both its positive charge and the size of its side chain. I find that KCNQ3-R230C is hard to close, but it is capable of being closed at strong negative voltages. I suggest that compounds that shift the voltage dependence of S4 activation to more positive potentials would promote gate closure and thus have therapeutic potential.
Collapse
Affiliation(s)
- Rene Barro-Soria
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
30
|
Lauxmann S, Verbeek NE, Liu Y, Zaichuk M, Müller S, Lemke JR, van Kempen MJ, Lerche H, Hedrich UB. Relationship of electrophysiological dysfunction and clinical severity in SCN2A
-related epilepsies. Hum Mutat 2018; 39:1942-1956. [DOI: 10.1002/humu.23619] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/09/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Stephan Lauxmann
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| | - Nienke E. Verbeek
- Department of Genetics; University Medical Centre Utrecht; Utrecht the Netherlands
| | - Yuanyuan Liu
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| | - Mariana Zaichuk
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| | - Stephan Müller
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| | - Johannes R. Lemke
- Institute of Human Genetics; University of Leipzig Hospitals and Clinics; Leipzig Germany
| | | | - Holger Lerche
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| | - Ulrike B.S. Hedrich
- Department of Neurology and Epileptology; Hertie Institute for Clinical Brain Research; University of Tuebingen; Tuebingen Germany
| |
Collapse
|
31
|
Kim EC, Zhang J, Pang W, Wang S, Lee KY, Cavaretta JP, Walters J, Procko E, Tsai NP, Chung HJ. Reduced axonal surface expression and phosphoinositide sensitivity in K v7 channels disrupts their function to inhibit neuronal excitability in Kcnq2 epileptic encephalopathy. Neurobiol Dis 2018; 118:76-93. [PMID: 30008368 DOI: 10.1016/j.nbd.2018.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/22/2018] [Accepted: 07/04/2018] [Indexed: 01/08/2023] Open
Abstract
Neuronal Kv7/KCNQ channels are voltage-gated potassium channels composed of Kv7.2/KCNQ2 and Kv7.3/KCNQ3 subunits. Enriched at the axonal membrane, they potently suppress neuronal excitability. De novo and inherited dominant mutations in Kv7.2 cause early onset epileptic encephalopathy characterized by drug resistant seizures and profound psychomotor delay. However, their precise pathogenic mechanisms remain elusive. Here, we investigated selected epileptic encephalopathy causing mutations in calmodulin (CaM)-binding helices A and B of Kv7.2. We discovered that R333W, K526N, and R532W mutations located peripheral to CaM contact sites decreased axonal surface expression of heteromeric channels although only R333W mutation reduced CaM binding to Kv7.2. These mutations also altered gating modulation by phosphatidylinositol 4,5-bisphosphate (PIP2), revealing novel PIP2 binding residues. While these mutations disrupted Kv7 function to suppress excitability, hyperexcitability was observed in neurons expressing Kv7.2-R532W that displayed severe impairment in voltage-dependent activation. The M518 V mutation at the CaM contact site in helix B caused most defects in Kv7 channels by severely reducing their CaM binding, K+ currents, and axonal surface expression. Interestingly, the M518 V mutation induced ubiquitination and accelerated proteasome-dependent degradation of Kv7.2, whereas the presence of Kv7.3 blocked this degradation. Furthermore, expression of Kv7.2-M518V increased neuronal death. Together, our results demonstrate that epileptic encephalopathy mutations in helices A and B of Kv7.2 cause abnormal Kv7 expression and function by disrupting Kv7.2 binding to CaM and/or modulation by PIP2. We propose that such multiple Kv7 channel defects could exert more severe impacts on neuronal excitability and health, and thus serve as pathogenic mechanisms underlying Kcnq2 epileptic encephalopathy.
Collapse
Affiliation(s)
- Eung Chang Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jiaren Zhang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Weilun Pang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shuwei Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - John P Cavaretta
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jennifer Walters
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Erik Procko
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
32
|
Oyrer J, Maljevic S, Scheffer IE, Berkovic SF, Petrou S, Reid CA. Ion Channels in Genetic Epilepsy: From Genes and Mechanisms to Disease-Targeted Therapies. Pharmacol Rev 2018; 70:142-173. [PMID: 29263209 DOI: 10.1124/pr.117.014456] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/02/2017] [Indexed: 12/19/2022] Open
Abstract
Epilepsy is a common and serious neurologic disease with a strong genetic component. Genetic studies have identified an increasing collection of disease-causing genes. The impact of these genetic discoveries is wide reaching-from precise diagnosis and classification of syndromes to the discovery and validation of new drug targets and the development of disease-targeted therapeutic strategies. About 25% of genes identified in epilepsy encode ion channels. Much of our understanding of disease mechanisms comes from work focused on this class of protein. In this study, we review the genetic, molecular, and physiologic evidence supporting the pathogenic role of a number of different voltage- and ligand-activated ion channels in genetic epilepsy. We also review proposed disease mechanisms for each ion channel and highlight targeted therapeutic strategies.
Collapse
Affiliation(s)
- Julia Oyrer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Ingrid E Scheffer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Samuel F Berkovic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| |
Collapse
|
33
|
Wild AR, Dell'Acqua ML. Potential for therapeutic targeting of AKAP signaling complexes in nervous system disorders. Pharmacol Ther 2017; 185:99-121. [PMID: 29262295 DOI: 10.1016/j.pharmthera.2017.12.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A common feature of neurological and neuropsychiatric disorders is a breakdown in the integrity of intracellular signal transduction pathways. Dysregulation of ion channels and receptors in the cell membrane and the enzymatic mediators that link them to intracellular effectors can lead to synaptic dysfunction and neuronal death. However, therapeutic targeting of these ubiquitous signaling elements can lead to off-target side effects due to their widespread expression in multiple systems of the body. A-kinase anchoring proteins (AKAPs) are multivalent scaffolding proteins that compartmentalize a diverse range of receptor and effector proteins to streamline signaling within nanodomain signalosomes. A number of essential neurological processes are known to critically depend on AKAP-directed signaling and an understanding of the role AKAPs play in nervous system disorders has emerged in recent years. Selective targeting of AKAP protein-protein interactions may be a means to uncouple pathologically active signaling pathways in neurological disorders with a greater degree of specificity. In this review we will discuss the role of AKAPs in both regulating normal nervous system function and dysfunction associated with disease, and the potential for therapeutic targeting of AKAP signaling complexes.
Collapse
Affiliation(s)
- Angela R Wild
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
34
|
Affiliation(s)
- Ananthanarayanan Kasinathan
- Department of Pediatrics, Pediatric Neurology and Neurodevelopment Unit, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Naveen Sankhyan
- Department of Pediatrics, Pediatric Neurology and Neurodevelopment Unit, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Pratibha Singhi
- Department of Pediatrics, Pediatric Neurology and Neurodevelopment Unit, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
35
|
Maljevic S, Reid CA, Petrou S. Models for discovery of targeted therapy in genetic epileptic encephalopathies. J Neurochem 2017; 143:30-48. [PMID: 28742937 DOI: 10.1111/jnc.14134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022]
Abstract
Epileptic encephalopathies are severe disorders emerging in the first days to years of life that commonly include refractory seizures, various types of movement disorders, and different levels of developmental delay. In recent years, many de novo occurring variants have been identified in individuals with these devastating disorders. To unravel disease mechanisms, the functional impact of detected variants associated with epileptic encephalopathies is investigated in a range of cellular and animal models. This review addresses efforts to advance and use such models to identify specific molecular and cellular targets for the development of novel therapies. We focus on ion channels as the best-studied group of epilepsy genes. Given the clinical and genetic heterogeneity of epileptic encephalopathy disorders, experimental models that can reflect this complexity are critical for the development of disease mechanisms-based targeted therapy. The convergence of technological advances in gene sequencing, stem cell biology, genome editing, and high throughput functional screening together with massive unmet clinical needs provides unprecedented opportunities and imperatives for precision medicine in epileptic encephalopathies.
Collapse
Affiliation(s)
- Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.,University of Melbourne, Melbourne, Australia
| |
Collapse
|
36
|
Gomis-Perez C, Soldovieri MV, Malo C, Ambrosino P, Taglialatela M, Areso P, Villarroel A. Differential Regulation of PI(4,5)P 2 Sensitivity of Kv7.2 and Kv7.3 Channels by Calmodulin. Front Mol Neurosci 2017; 10:117. [PMID: 28507506 PMCID: PMC5410570 DOI: 10.3389/fnmol.2017.00117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 11/19/2022] Open
Abstract
HIGHLIGHTS- Calmodulin-dependent Kv7.2 current density without the need of binding calcium. - Kv7.2 current density increase is accompanied with resistance to PI(4,5)P2 depletion. - Kv7.3 current density is insensitive to calmodulin elevation. - Kv7.3 is more sensitive to PI(4,5)P2 depletion in the presence of calmodulin. - Apo-calmodulin influences PI(4,5)P2 dependence in a subunit specific manner.
The identification and understanding of critical factors regulating M-current functional density, whose main components are Kv7.2 and Kv7.3 subunits, has profound pathophysiological impact given the important role of the M-current in neuronal excitability control. We report the increase in current density of Kv7.2 channels by calmodulin (CaM) and by a mutant CaM unable to bind Ca2+ (CaM1234) revealing that this potentiation is calcium independent. Furthermore, after co-expressing a CaM binding protein (CaM sponge) to reduce CaM cellular availability, Kv7.2 current density was reduced. Current inhibition after transient depletion of the essential Kv7 co-factor phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) by activating Danio rerio voltage sensitive phosphatase (DrVSP) was blunted by co-expressing CaM1234 or the CaM sponge. In addition, CaM-dependent potentiation was occluded by tonic elevation of PI(4,5)P2 levels by PI(4)P5-kinase (PIP5K) expression. In contrast to the effect on homomeric Kv7.2 channels, CaM1234 failed to potentiate heteromeric Kv7.2/3 or homomeric Kv7.3 channels. Sensitivity to PI(4,5)P2 depletion of Kv7.2/3 channels was increased after expression of CaM1234 or the CaM sponge, while that of homomeric Kv7.3 was unaltered. Altogether, the data reveal that apo-CaM influences PI(4,5)P2 dependence of Kv7.2, Kv7.2/3, and of Kv7.3 channels in a subunit specific manner.
Collapse
Affiliation(s)
- Carolina Gomis-Perez
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| | - Maria V Soldovieri
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Covadonga Malo
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| | - Paolo Ambrosino
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Maurizio Taglialatela
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy.,Department of Neuroscience, University of Naples "Federico II,"Naples, Italy
| | - Pilar Areso
- Department Farmacología, UPV/EHU, Universidad del País VascoLeioa, Spain
| | - Alvaro Villarroel
- Biofisika Institutua, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHULeioa, Spain
| |
Collapse
|
37
|
Rinker JA, Mulholland PJ. Promising pharmacogenetic targets for treating alcohol use disorder: evidence from preclinical models. Pharmacogenomics 2017; 18:555-570. [PMID: 28346058 DOI: 10.2217/pgs-2016-0193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inherited genetic variants contribute to risk factors for developing an alcohol use disorder, and polymorphisms may inform precision medicine strategies for treating alcohol addiction. Targeting genetic mutations linked to alcohol phenotypes has provided promising initial evidence for reducing relapse rates in alcoholics. Although successful in some studies, there are conflicting findings and the reports of adverse effects may ultimately limit their clinical utility, suggesting that novel pharmacogenetic targets are necessary to advance precision medicine approaches. Here, we describe promising novel genetic variants derived from preclinical models of alcohol consumption and dependence that may uncover disease mechanisms that drive uncontrolled drinking and identify novel pharmacogenetic targets that facilitate therapeutic intervention for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
38
|
Xiong W, Zhou D. Progress in unraveling the genetic etiology of rolandic epilepsy. Seizure 2017; 47:99-104. [PMID: 28351718 DOI: 10.1016/j.seizure.2017.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
Rolandic epilepsy (RE), or benign epilepsy of childhood with centrotemporal spikes (BECT), is the most frequent idiopathic partial epilepsy syndrome of childhood, where the "idiopathic" implies a genetic predisposition. Although RE has long been presumed to have a genetic component, clinical and genetic studies have shown a complex inheritance pattern. Furthermore, the underlying major genetic influence in RE has been challenged by recent reports of twin studies. Meanwhile, many genes or loci have been shown to be associated the RE/atypical RE (ARE) spectrum, with a higher frequency of causative variants in ARE. However, a full understanding of the genetic basis in the more common forms of the RE spectrum remains elusive.
Collapse
Affiliation(s)
- Weixi Xiong
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dong Zhou
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
39
|
Rinker JA, Fulmer DB, Trantham-Davidson H, Smith ML, Williams RW, Lopez MF, Randall PK, Chandler LJ, Miles MF, Becker HC, Mulholland PJ. Differential potassium channel gene regulation in BXD mice reveals novel targets for pharmacogenetic therapies to reduce heavy alcohol drinking. Alcohol 2017; 58:33-45. [PMID: 27432260 DOI: 10.1016/j.alcohol.2016.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/12/2016] [Accepted: 05/03/2016] [Indexed: 12/22/2022]
Abstract
Alcohol (ethanol) dependence is a chronic relapsing brain disorder partially influenced by genetics and characterized by an inability to regulate harmful levels of drinking. Emerging evidence has linked genes that encode KV7, KIR, and KCa2 K+ channels with variation in alcohol-related behaviors in rodents and humans. This led us to experimentally test relations between K+ channel genes and escalation of drinking in a chronic-intermittent ethanol (CIE) exposure model of dependence in BXD recombinant inbred strains of mice. Transcript levels for K+ channel genes in the prefrontal cortex (PFC) and nucleus accumbens (NAc) covary with voluntary ethanol drinking in a non-dependent cohort. Transcripts that encode KV7 channels covary negatively with drinking in non-dependent BXD strains. Using a pharmacological approach to validate the genetic findings, C57BL/6J mice were allowed intermittent access to ethanol to establish baseline consumption before they were treated with retigabine, an FDA-approved KV7 channel positive modulator. Systemic administration significantly reduced drinking, and consistent with previous evidence, retigabine was more effective at reducing voluntary consumption in high-drinking than low-drinking subjects. We evaluated the specific K+ channel genes that were most sensitive to CIE exposure and identified a gene subset in the NAc and PFC that were dysregulated in the alcohol-dependent BXD cohort. CIE-induced modulation of nine genes in the NAc and six genes in the PFC covaried well with the changes in drinking induced by ethanol dependence. Here we identified novel candidate genes in the NAc and PFC that are regulated by ethanol dependence and correlate with voluntary drinking in non-dependent and dependent BXD mice. The findings that Kcnq expression correlates with drinking and that retigabine reduces consumption suggest that KV7 channels could be pharmacogenetic targets to treat individuals with alcohol addiction.
Collapse
|
40
|
Malik R, Mehta P, Srivastava S, Choudhary BS, Sharma M. Pharmacophore modeling, 3D-QSAR, and in silico ADME prediction of N-pyridyl and pyrimidine benzamides as potent antiepileptic agents. J Recept Signal Transduct Res 2016; 37:259-266. [PMID: 27607834 DOI: 10.1080/10799893.2016.1217883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Biological mechanism attributing mutations in KCNQ2/Q3 results in benign familial neonatal epilepsy (BFNE), a rare form of epilepsy and thus neglected. It offers a potential target for antiepileptic drug discovery. In the present work, a pharmacophore-based 3D-QSAR model was generated for a series of N-pyridyl and pyrimidine benzamides possessing KCNQ2/Q3 opening activity. The pharmacophore model generated contains one hydrogen bond donor (D), one hydrophobic (H), and two aromatic rings (R). They are the crucial molecular write-up detailing predicted binding efficacy of high affinity and low affinity ligands for KCNQ2/Q3 opening activity. Furthermore, it has been validated by using a biological correlation between pharmacophore hypothesis-based 3D-QSAR variables and functional fingerprints of openers responsible for the receptor binding and also by docking of these benzamides into the validated homology model. Excellent statistical computational tools of QSAR model such as good correlation coefficient (R2 > 0.80), higher F value (F > 39), and excellent predictive power (Q2 > 0.7) with low standard deviation (SD <0.3) strongly suggest that the developed model could be used for prediction of antiepileptic activity of newer analogs. A preliminary pharmacokinetic profile of these derivatives was also performed on the basis of QikProp predictions.
Collapse
Affiliation(s)
- Ruchi Malik
- a Department of Pharmacy, School of Chemical Sciences and Pharmacy , Central University of Rajasthan , Kishangarh, Ajmer , Rajasthan , India
| | - Pakhuri Mehta
- a Department of Pharmacy, School of Chemical Sciences and Pharmacy , Central University of Rajasthan , Kishangarh, Ajmer , Rajasthan , India
| | - Shubham Srivastava
- a Department of Pharmacy, School of Chemical Sciences and Pharmacy , Central University of Rajasthan , Kishangarh, Ajmer , Rajasthan , India
| | - Bhanwar Singh Choudhary
- a Department of Pharmacy, School of Chemical Sciences and Pharmacy , Central University of Rajasthan , Kishangarh, Ajmer , Rajasthan , India
| | - Manish Sharma
- b School of Pharmacy , Maharishi Markandeshwar University , Sadopur, Ambala , Haryana , India
| |
Collapse
|
41
|
Thiffault I, Speca DJ, Austin DC, Cobb MM, Eum KS, Safina NP, Grote L, Farrow EG, Miller N, Soden S, Kingsmore SF, Trimmer JS, Saunders CJ, Sack JT. A novel epileptic encephalopathy mutation in KCNB1 disrupts Kv2.1 ion selectivity, expression, and localization. ACTA ACUST UNITED AC 2016; 146:399-410. [PMID: 26503721 PMCID: PMC4621747 DOI: 10.1085/jgp.201511444] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A missense mutation in the pore-forming α subunit of a delayed rectifier Kv channel is associated with epileptic encephalopathy, alters the cation selectivity of voltage-gated currents, and disrupts channel expression and localization. The epileptic encephalopathies are a group of highly heterogeneous genetic disorders. The majority of disease-causing mutations alter genes encoding voltage-gated ion channels, neurotransmitter receptors, or synaptic proteins. We have identified a novel de novo pathogenic K+ channel variant in an idiopathic epileptic encephalopathy family. Here, we report the effects of this mutation on channel function and heterologous expression in cell lines. We present a case report of infantile epileptic encephalopathy in a young girl, and trio-exome sequencing to determine the genetic etiology of her disorder. The patient was heterozygous for a de novo missense variant in the coding region of the KCNB1 gene, c.1133T>C. The variant encodes a V378A mutation in the α subunit of the Kv2.1 voltage-gated K+ channel, which is expressed at high levels in central neurons and is an important regulator of neuronal excitability. We found that expression of the V378A variant results in voltage-activated currents that are sensitive to the selective Kv2 channel blocker guangxitoxin-1E. These voltage-activated Kv2.1 V378A currents were nonselective among monovalent cations. Striking cell background–dependent differences in expression and subcellular localization of the V378A mutation were observed in heterologous cells. Further, coexpression of V378A subunits and wild-type Kv2.1 subunits reciprocally affects their respective trafficking characteristics. A recent study reported epileptic encephalopathy-linked missense variants that render Kv2.1 a tonically activated, nonselective cation channel that is not voltage activated. Our findings strengthen the correlation between mutations that result in loss of Kv2.1 ion selectivity and development of epileptic encephalopathy. However, the strong voltage sensitivity of currents from the V378A mutant indicates that the loss of voltage-sensitive gating seen in all other reported disease mutants is not required for an epileptic encephalopathy phenotype. In addition to electrophysiological differences, we suggest that defects in expression and subcellular localization of Kv2.1 V378A channels could contribute to the pathophysiology of this KCNB1 variant.
Collapse
Affiliation(s)
- Isabelle Thiffault
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108
| | - David J Speca
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| | - Daniel C Austin
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| | - Melanie M Cobb
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| | - Kenneth S Eum
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| | - Nicole P Safina
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108
| | - Lauren Grote
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108
| | - Emily G Farrow
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108
| | - Neil Miller
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108
| | - Sarah Soden
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108
| | - Stephen F Kingsmore
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616 Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| | - Carol J Saunders
- Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 Center for Pediatric Genomic Medicine, Department of Pathology and Laboratory Medicine, and Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108 University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108
| | - Jon T Sack
- Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616 Department of Neurobiology, Physiology and Behavior, Department of Physiology and Membrane Biology, and Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616
| |
Collapse
|
42
|
Maljevic S, Vejzovic S, Bernhard MK, Bertsche A, Weise S, Döcker M, Lerche H, Lemke JR, Merkenschlager A, Syrbe S. Novel KCNQ3 Mutation in a Large Family with Benign Familial Neonatal Epilepsy: A Rare Cause of Neonatal Seizures. Mol Syndromol 2016; 7:189-196. [PMID: 27781029 DOI: 10.1159/000447461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Benign familial neonatal seizures (BFNS) present a rare familial epilepsy syndrome caused by genetic alterations in the voltage-gated potassium channels Kv7.2 and Kv7.3, encoded by KCNQ2 and KCNQ3. While most BFNS families carry alterations in KCNQ2, mutations in KCNQ3 appear to be less common. Here, we describe a family with 6 individuals presenting with neonatal focal and generalized seizures. Genetic testing revealed a novel KCNQ3 variant, c.835G>T, cosegregating with seizures in 4 tested individuals. This variant results in a substitution of the highly conserved amino acid valine localized within the pore-forming transmembrane segment S5 (p.V279F). Functional investigations in Xenopus laevis oocytes revealed a loss of function, which supports p.V279F as a pathogenic mutation. When p.V279F was coexpressed with the wild-type (WT) Kv7.2 subunits, the resulting potassium currents were about 10-fold reduced compared to the WT Kv7.3 and Kv7.2 coexpression. Genotype-phenotype correlation shows an incomplete penetrance of p.V279F. Response to antiepileptic treatment was variable, but evaluation of treatment response remained challenging due to the self-limiting character of the disease. The identification of the pathogenic variant helped to avoid unnecessary investigations in affected family members and allowed guided therapy.
Collapse
Affiliation(s)
- Snezana Maljevic
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Heidelberg, Germany; The Florey Institute of Neuroscience and Mental Health, Melbourne, Vic., Australia
| | - Sabina Vejzovic
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Heidelberg, Germany
| | - Matthias K Bernhard
- Department of Women and Child Health, Hospital for Children and Adolescents, Heidelberg, Germany
| | - Astrid Bertsche
- Department of Women and Child Health, Hospital for Children and Adolescents, Heidelberg, Germany
| | - Sebastian Weise
- Department of Women and Child Health, Hospital for Children and Adolescents, Heidelberg, Germany
| | - Miriam Döcker
- CeGaT GmbH, Tübingen, University Hospital Heidelberg, Heidelberg, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Heidelberg, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig, Germany
| | - Andreas Merkenschlager
- Department of Women and Child Health, Hospital for Children and Adolescents, Heidelberg, Germany
| | - Steffen Syrbe
- Department of Women and Child Health, Hospital for Children and Adolescents, Heidelberg, Germany; Division of Child Neurology and Inherited Metabolic Diseases, Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
43
|
Verrotti A, Zara F, Minetti C, Striano P. Novel treatment perspectives from advances in understanding of genetic epilepsy syndromes. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1167594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Striano P, Vari MS, Mazzocchetti C, Verrotti A, Zara F. Management of genetic epilepsies: From empirical treatment to precision medicine. Pharmacol Res 2016; 107:426-429. [PMID: 27080588 DOI: 10.1016/j.phrs.2016.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 04/07/2016] [Accepted: 04/09/2016] [Indexed: 01/17/2023]
Abstract
Despite the over 20 antiepileptic drugs (AEDs) now licensed for epilepsy treatment, seizures can be effectively controlled in about ∼70% of patients. Thus, epilepsy treatment is still challenging in about one third of patients and this may lead to a severe medically, physically, and socially disabling condition. However, there is clear evidence of heterogeneity of response to existing AEDs and a significant unmet need for effective intervention. A number of studies have shown that polymorphisms may influence the poor or inadequate therapeutic response as well as the occurrence of adverse effects. In addition, the new frontier of genomic technologies, including chromosome microarrays and next-generation sequencing, improved our understanding of the genetic architecture of epilepsies. Recent findings in some genetic epilepsy syndromes provide insights into mechanisms of epileptogenesis, unrevealing the role of a number of genes with different functions, such as ion channels, proteins associated to the vesical synaptic cycle or involved in energy metabolism. The rapid progress of high-throughput genomic sequencing and corresponding analysis tools in molecular diagnosis are revolutionizing the practice and it is a fact that for some monogenic epilepsies the molecular confirmation may influence the choice of the treatment. Moreover, the novel genetic methods, that are able to analyze all known genes at a reasonable price, are of paramount importance to discover novel therapeutic avenues and individualized (or precision) medicine.
Collapse
Affiliation(s)
- Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, G. Gaslini Institute, Genova, Italy.
| | - Maria Stella Vari
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, G. Gaslini Institute, Genova, Italy
| | | | - Alberto Verrotti
- Department of Pediatrics, University of L' Aquila, LAquila, Italy
| | - Federico Zara
- Laboratory of Neurosciences and Neurogenetics, Department of Head and Neck Diseases, G. Gaslini Institute, Genova, Italy
| |
Collapse
|
45
|
Devaux J, Abidi A, Roubertie A, Molinari F, Becq H, Lacoste C, Villard L, Milh M, Aniksztejn L. A Kv7.2 mutation associated with early onset epileptic encephalopathy with suppression-burst enhances Kv7/M channel activity. Epilepsia 2016; 57:e87-93. [PMID: 27030113 DOI: 10.1111/epi.13366] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2016] [Indexed: 02/04/2023]
Abstract
Mutations in the KCNQ2 gene encoding the voltage-gated potassium channel subunit Kv7.2 cause early onset epileptic encephalopathy (EOEE). Most mutations have been shown to induce a loss of function or to affect the subcellular distribution of Kv7 channels in neurons. Herein, we investigated functional consequences and subcellular distribution of the p.V175L mutation of Kv7.2 (Kv7.2(V175L) ) found in a patient presenting EOEE. We observed that the mutation produced a 25-40 mV hyperpolarizing shift of the conductance-voltage relationship of both the homomeric Kv7.2(V175L) and heteromeric Kv7.2(V175L) /Kv7.3 channels compared to wild-type channels and a 10 mV hyperpolarizing shift of Kv7.2(V175L) /Kv7.2/Kv7.3 channels in a 1:1:2 ratio mimicking the patient situation. Mutant channels also displayed faster activation kinetics and an increased current density that was prevented by 1 μm linopirdine. The p.V175L mutation did not affect the protein expression of Kv7 channels and its localization at the axon initial segment. We conclude that p.V175L is a gain of function mutation. This confirms previous observations showing that mutations having opposite consequences on M channels can produce EOEE. These findings alert us that drugs aiming to increase Kv7 channel activity might have adverse effects in EOEE in the case of gain-of-function variants.
Collapse
Affiliation(s)
- Jérôme Devaux
- CNRS, Aix-Marseille University, CRN2M-UMR7286, Marseille, France
| | - Affef Abidi
- GMGF, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 910, Marseille, France
| | - Agathe Roubertie
- Pediatric Neurology Department, Montpellier University Hospital, Montpellier, France.,INSERM U1051, INM Montpellier, Montpellier, France
| | - Florence Molinari
- Mediterranean Neurobiology Institute INMED, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 901, Marseille, France
| | - Hélène Becq
- Mediterranean Neurobiology Institute INMED, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 901, Marseille, France
| | - Caroline Lacoste
- GMGF, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 910, Marseille, France.,Timone Children Hospital, Pediatric Neurology Department, APHM, Marseille, France
| | - Laurent Villard
- GMGF, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 910, Marseille, France
| | - Mathieu Milh
- GMGF, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 910, Marseille, France.,Timone Children Hospital, Pediatric Neurology Department, APHM, Marseille, France
| | - Laurent Aniksztejn
- Mediterranean Neurobiology Institute INMED, Aix-Marseille University, Marseille, France.,INSERM, UMR_S 901, Marseille, France
| |
Collapse
|
46
|
Zhu WY, Jiang P, He X, Cao LJ, Zhang LH, Dang RL, Tang MM, Xue Y, Li HD. Contribution of NRG1 Gene Polymorphisms in Temporal Lobe Epilepsy. J Child Neurol 2016; 31:271-6. [PMID: 26071373 DOI: 10.1177/0883073815589757] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/11/2015] [Indexed: 02/04/2023]
Abstract
The purpose of the present study was to investigate the possible association between temporal lobe epilepsy and NRG1 gene polymorphisms. A total of 73 patients and 69 controls were involved in this study. Genomic DNAs from the patients and controls were genotyped by polymerase chain reaction-ligase detection reaction method. There was an association of rs35753505 (T>C) with temporal lobe epilepsy (χ(2) = 6.730, P = .035). The frequency of risk allele C of rs35753505 was significantly higher (69.9%) in patients compared to controls (55.8%) (χ(2) = 6.023, P = .014). Interestingly, the significant difference of NRG1 genotype and allele frequency only existed among males, but not females. In addition, no statistically significant association was found between rs6994992, rs62510682 polymorphisms, and temporal lobe epilepsy. These data indicate that rs35753505 of NRG1 plays an important role in conferring susceptibility to the temporal lobe epilepsy in a Chinese Han population.
Collapse
Affiliation(s)
- Wen-Ye Zhu
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Pei Jiang
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Xin He
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Ling-Juan Cao
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Li-Hong Zhang
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Rui-Li Dang
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Mi-Mi Tang
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Ying Xue
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China School of Pharmaceutical Science, Central South University, Changsha, Hunan Province, China
| | - Huan-De Li
- Institute of Clinical Pharmacy & Pharmacology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
47
|
Ohya S, Kito H, Hatano N, Muraki K. Recent advances in therapeutic strategies that focus on the regulation of ion channel expression. Pharmacol Ther 2016; 160:11-43. [PMID: 26896566 DOI: 10.1016/j.pharmthera.2016.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of different ion channel types are involved in cell signaling networks, and homeostatic regulatory mechanisms contribute to the control of ion channel expression. Profiling of global gene expression using microarray technology has recently provided novel insights into the molecular mechanisms underlying the homeostatic and pathological control of ion channel expression. It has demonstrated that the dysregulation of ion channel expression is associated with the pathogenesis of neural, cardiovascular, and immune diseases as well as cancers. In addition to the transcriptional, translational, and post-translational regulation of ion channels, potentially important evidence on the mechanisms controlling ion channel expression has recently been accumulated. The regulation of alternative pre-mRNA splicing is therefore a novel therapeutic strategy for the treatment of dominant-negative splicing disorders. Epigenetic modification plays a key role in various pathological conditions through the regulation of pluripotency genes. Inhibitors of pre-mRNA splicing and histone deacetyalase/methyltransferase have potential as potent therapeutic drugs for cancers and autoimmune and inflammatory diseases. Moreover, membrane-anchoring proteins, lysosomal and proteasomal degradation-related molecules, auxiliary subunits, and pharmacological agents alter the protein folding, membrane trafficking, and post-translational modifications of ion channels, and are linked to expression-defect channelopathies. In this review, we focused on recent insights into the transcriptional, spliceosomal, epigenetic, and proteasomal regulation of ion channel expression: Ca(2+) channels (TRPC/TRPV/TRPM/TRPA/Orai), K(+) channels (voltage-gated, KV/Ca(2+)-activated, KCa/two-pore domain, K2P/inward-rectifier, Kir), and Ca(2+)-activated Cl(-) channels (TMEM16A/TMEM16B). Furthermore, this review highlights expression of these ion channels in expression-defect channelopathies.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan.
| |
Collapse
|
48
|
Kuzmenkov AI, Grishin EV, Vassilevski AA. Diversity of Potassium Channel Ligands: Focus on Scorpion Toxins. BIOCHEMISTRY (MOSCOW) 2016; 80:1764-99. [DOI: 10.1134/s0006297915130118] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Alberdi A, Gomis-Perez C, Bernardo-Seisdedos G, Alaimo A, Malo C, Aldaregia J, Lopez-Robles C, Areso P, Butz E, Wahl-Schott C, Villarroel A. Uncoupling PIP2-calmodulin regulation of Kv7.2 channels by an assembly destabilizing epileptogenic mutation. J Cell Sci 2015; 128:4014-23. [PMID: 26359296 DOI: 10.1242/jcs.176420] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/09/2015] [Indexed: 01/15/2023] Open
Abstract
We show that the combination of an intracellular bi-partite calmodulin (CaM)-binding site and a distant assembly region affect how an ion channel is regulated by a membrane lipid. Our data reveal that regulation by phosphatidylinositol(4,5)bisphosphate (PIP2) and stabilization of assembled Kv7.2 subunits by intracellular coiled-coil regions far from the membrane are coupled molecular processes. Live-cell fluorescence energy transfer measurements and direct binding studies indicate that remote coiled-coil formation creates conditions for different CaM interaction modes, each conferring different PIP2 dependency to Kv7.2 channels. Disruption of coiled-coil formation by epilepsy-causing mutation decreases apparent CaM-binding affinity and interrupts CaM influence on PIP2 sensitivity.
Collapse
Affiliation(s)
- Araitz Alberdi
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Carolina Gomis-Perez
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Ganeko Bernardo-Seisdedos
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Alessandro Alaimo
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Covadonga Malo
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Juncal Aldaregia
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Carlos Lopez-Robles
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Pilar Areso
- Departament de Farmacología, UPV/EHU, Universidad del País Vasco, Barrio Sarriena s/n, Leioa 48940, Spain
| | - Elisabeth Butz
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität, München 81377, Germany
| | - Christian Wahl-Schott
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität, München 81377, Germany
| | - Alvaro Villarroel
- Unidad de Biofísica, Consejo Superior de Investigaciones Científicas, CSIC, UPV/EHU, Barrio Sarriena s/n, Leioa 48940, Spain
| |
Collapse
|
50
|
Abstract
As the genetic etiologies of an expanding number of epilepsy syndromes are revealed, the complexity of the phenotype genotype correlation increases. As our review will show, multiple gene mutations cause different epilepsy syndromes, making identification of the specific mutation increasingly more important for prognostication and often more directed treatment. Examples of that include the need to avoid specific drugs in Dravet syndrome and the ongoing investigations of the potential use of new directed therapies such as retigabine in KCNQ2-related epilepsies, quinidine in KCNT1-related epilepsies, and memantine in GRIN2A-related epilepsies.
Collapse
Affiliation(s)
- Abeer J Hani
- Division of Pediatric Neurology, Department of Pediatrics, Duke Children's Hospital and Health Center, Suite T0913J, 2301 Erwin Road, Durham, NC 27710, USA
| | - Husam M Mikati
- Center of Human Genome Variation, LSRC, Duke University School of Medicine, 201 Trent Drive, Durham, NC 27710, USA
| | - Mohamad A Mikati
- Division of Pediatric Neurology, Department of Pediatrics, Duke Children's Hospital and Health Center, Suite T0913J, 2301 Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|