1
|
Yang Z, Zheng Y, Ma D, Wang L, Zhang J, Song T, Wang Y, Zhang Y, Nan F, Su N, Gao Z, Guo J. Phosphatidylinositol 4,5-bisphosphate activation mechanism of human KCNQ5. Proc Natl Acad Sci U S A 2025; 122:e2416738122. [PMID: 40172963 PMCID: PMC12002238 DOI: 10.1073/pnas.2416738122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Abstract
The human voltage-gated potassium channels KCNQ2, KCNQ3, and KCNQ5 can form homo- and heterotetrameric channels that are responsible for generating the neuronal M current and maintaining the membrane potential stable. Activation of KCNQ channels requires both the depolarization of membrane potential and phosphatidylinositol 4,5-bisphosphate (PIP2). Here, we report cryoelectron microscopy structures of the human KCNQ5-calmodulin (CaM) complex in the apo, PIP2-bound, and both PIP2- and the activator HN37-bound states in either a closed or an open conformation. In the closed conformation, a PIP2 molecule binds in the middle of the groove between two adjacent voltage-sensing domains (VSDs), whereas in the open conformation, one additional PIP2 binds to the interface of VSD and the pore domain, accompanying structural rearrangement of the cytosolic domain of KCNQ and CaM. The structures, along with electrophysiology analyses, reveal the two different binding modes of PIP2 and elucidate the PIP2 activation mechanism of KCNQ5.
Collapse
Affiliation(s)
- Zhenni Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang311100, China
| | - Yueming Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Demin Ma
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang311100, China
| | - Long Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Jiatong Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang311100, China
| | - Tiefeng Song
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang310058, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang310058, China
| | - Yan Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang311100, China
| | - Fajun Nan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Nannan Su
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang322000, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong528437, China
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang311100, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang311121, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang310058, China
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang310058, China
| |
Collapse
|
2
|
Mao M, Jancovski N, Kushner Y, Teasdale L, Truong P, Zhou K, Reid S, Jia L, Aung YH, Li M, Reid CA, Byars S, Scheffer I, Petrou S, Maljevic S. Developmental dysfunction in a preclinical model of Kcnq2 developmental and epileptic encephalopathy. Neurobiol Dis 2025; 205:106782. [PMID: 39733957 DOI: 10.1016/j.nbd.2024.106782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/23/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Developmental and epileptic encephalopathies (DEE) are rare but severe neurodevelopmental disorders characterised by early-onset seizures often combined with developmental delay, behavioural and cognitive deficits. Treatment for DEEs is currently limited to seizure control and provides no benefits to the patients' developmental and cognitive outcomes. Genetic variants are the most common cause of DEE with KCNQ2 being one of the most frequently identified disease-causing genes. KCNQ2 encodes a voltage-gated potassium channel KV7.2 widely expressed in the central nervous system and critically involved in the regulation of neuronal excitability. In this study, we aimed to characterise a KCNQ2 variant (K556E) found in a female patient with DEE using a heterologous expression system and a knock-in mouse model. METHODS Wild-type KCNQ2 or K556E variant were expressed in Chinese Hamster Ovary (CHO) cells (with or without KCNQ3) and their biophysical properties assessed using patch clamp recordings. We further engineered a new Kcnq2 DEE mouse model (K557E) based on the K556E variant and characterised it using behavioural, electrophysiological, and transcriptome analysis. RESULTS A mild loss of function was observed only when the mutant channel was co-expressed with KCNQ3 in the heterologous system. The heterozygous knock-in mice showed a reduced survival rate and increased susceptibility to induced seizures. Electrophysiology recordings in brain slices revealed a hyperexcitable phenotype for cortical layer 2/3 pyramidal neurons with retigabine (KV7 channel opener) able to rescue both the increased sensitivity to chemically-induced seizures in vivo and neuronal excitability ex vivo. Whole-brain RNA sequencing revealed numerous differentially expressed genes and biological pathways pointing at dysregulation of early developmental processes. CONCLUSIONS Our study reports on a novel Kcnq2 DEE mouse model recapitulating aspects of the disease phenotype with the electrophysiological and transcriptome analysis providing insights into KCNQ2 DEE mechanisms that can be leveraged for future therapy development.
Collapse
Affiliation(s)
- Miaomiao Mao
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Nikola Jancovski
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Yafit Kushner
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Lucas Teasdale
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Phan Truong
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Kun Zhou
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Samuel Reid
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Linghan Jia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Ye Htet Aung
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Melody Li
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Sean Byars
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Ingrid Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia; Praxis Precision Medicine Inc, Cambridge, MA 02142, USA.
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.
| |
Collapse
|
3
|
Lemke J, Gollasch M, Tsvetkov D, Schulig L. Advances in the design and development of chemical modulators of the voltage-gated potassium channels K V7.4 and K V7.5. Expert Opin Drug Discov 2025; 20:47-62. [PMID: 39627683 DOI: 10.1080/17460441.2024.2438226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/20/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Hypertension remains a major public health concern, with significant morbidity and mortality worldwide. Despite the availability of various antihypertensive medications, blood pressure control remains suboptimal in many individuals. During the last decades, KV7.4 and KV7.5, which were already known from the view of neuronal regulation, emerged as possible important players in the regulation of vascular tone and blood pressure. AREAS COVERED This review covers physiological functions and current advancements in the development of KV7.4 and KV7.5 channel modulators. The authors highlight the structural elements likely to be important for the future design of KV7 subtype-selective modulators, underscoring their potential as an innovative hypertension treatment. EXPERT OPINION Extensive research has been focused on targeting neuronal KV7.2 and KV7.3 channels, while KV7.4 and KV7.5 attracted less attention. Many of the developed compounds represent derivatives of flupirtine or retigabine, whereby subtype channel selectivity has only been demonstrated for a handful of individual compounds. Novel substances address additional sites within the binding pocket by incorporating new functional groups. A comprehensive and systematic evaluation of a compound set with significant subtype selectivity should be performed. The discovery of new highly active, less toxic, and selective compounds, therefore, remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Jana Lemke
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Abbott GW, Manville RW. Discovery of a potent, Kv7.3-selective potassium channel opener from a Polynesian traditional botanical anticonvulsant. Commun Chem 2024; 7:233. [PMID: 39390220 PMCID: PMC11467302 DOI: 10.1038/s42004-024-01318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Plants remain an important source of biologically active small molecules with high therapeutic potential. The voltage-gated potassium (Kv) channel formed by Kv7.2/3 (KCNQ2/3) heteromers is a major target for anticonvulsant drug development. Here, we screened 1444 extracts primarily from plants collected in California and the US Virgin Islands, for their ability to activate Kv7.2/3 but not inhibit Kv1.3, to select against tannic acid being the active component. We validated the 7 strongest hits, identified Thespesia populnea (miro, milo, portia tree) as the most promising, then discovered its primary active metabolite to be gentisic acid (GA). GA highly potently activated Kv7.2/3 (EC50, 2.8 nM). GA is, uniquely to our knowledge, 100% selective for Kv7.3 versus other Kv7 homomers; it requires S5 residue Kv7.3-W265 for Kv7.2/3 activation, and it ameliorates pentylenetetrazole-induced seizures in mice. Structure-activity studies revealed that the FDA-approved vasoprotective drug calcium dobesilate, a GA analog, is a previously unrecognized Kv7.2/3 channel opener. Also an active aspirin metabolite, GA provides a molecular rationale for the use of T. populnea as an anticonvulsant in Polynesian indigenous medicine and presents novel pharmacological prospects for potent, isoform-selective, therapeutic Kv7 channel activation.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
5
|
Rönnelid O, Elinder F. Carboxyl-group compounds activate voltage-gated potassium channels via a distinct mechanism. J Gen Physiol 2024; 156:e202313516. [PMID: 38832889 PMCID: PMC11148469 DOI: 10.1085/jgp.202313516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/14/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024] Open
Abstract
Voltage-gated ion channels are responsible for the electrical excitability of neurons and cardiomyocytes. Thus, they are obvious targets for pharmaceuticals aimed to modulate excitability. Compounds activating voltage-gated potassium (KV) channels are expected to reduce excitability. To search for new KV-channel activators, we performed a high-throughput screen of 10,000 compounds on a specially designed Shaker KV channel. Here, we report on a large family of channel-activating compounds with a carboxyl (COOH) group as the common motif. The most potent COOH activators are lipophilic (4 < LogP <7) and are suggested to bind at the interface between the lipid bilayer and the channel's positively charged voltage sensor. The negatively charged form of the COOH-group compounds is suggested to open the channel by electrostatically pulling the voltage sensor to an activated state. Several of the COOH-group compounds also activate the therapeutically important KV7.2/7.3 channel and can thus potentially be developed into antiseizure drugs. The COOH-group compounds identified in this study are suggested to act via the same site and mechanism of action as previously studied COOH-group compounds, such as polyunsaturated fatty acids and resin acids, but distinct from sites for several other types of potassium channel-activating compounds.
Collapse
Affiliation(s)
- Olle Rönnelid
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Elinder
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Science for Life Laboratory, Linköping University, Linköping, Sweden
| |
Collapse
|
6
|
Graziano B, Wang L, White OR, Kaplan DH, Fernandez-Abascal J, Bianchi L. Glial KCNQ K + channels control neuronal output by regulating GABA release from glia in C. elegans. Neuron 2024; 112:1832-1847.e7. [PMID: 38460523 PMCID: PMC11156561 DOI: 10.1016/j.neuron.2024.02.013] [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: 10/04/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/11/2024]
Abstract
KCNQs are voltage-gated K+ channels that control neuronal excitability and are mutated in epilepsy and autism spectrum disorder (ASD). KCNQs have been extensively studied in neurons, but their function in glia is unknown. Using voltage, calcium, and GABA imaging, optogenetics, and behavioral assays, we show here for the first time in Caenorhabditis elegans (C. elegans) that glial KCNQ channels control neuronal excitability by mediating GABA release from glia via regulation of the function of L-type voltage-gated Ca2+ channels. Further, we show that human KCNQ channels have the same role when expressed in nematode glia, underscoring conservation of function across species. Finally, we show that pathogenic loss-of-function and gain-of-function human KCNQ2 mutations alter glia-to-neuron GABA signaling in distinct ways and that the KCNQ channel opener retigabine exerts rescuing effects. This work identifies glial KCNQ channels as key regulators of neuronal excitability via control of GABA release from glia.
Collapse
Affiliation(s)
- Bianca Graziano
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lei Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Olivia R White
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daryn H Kaplan
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
7
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596557. [PMID: 38853899 PMCID: PMC11160764 DOI: 10.1101/2024.05.29.596557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit, receiving strong input from the indirect pathway and regulating a variety of functions, including locomotor output and habit formation. We recently showed that it also acts as a gatekeeper of cocaine-induced behavioral plasticity, as inhibition of parvalbumin-positive cells in the GPe (GPe PV ) prevents the development of cocaine-induced reward and sensitization. However, the molecular and circuit mechanisms underlying this function are unknown. Here we show that GPe PV cells control cocaine reward and sensitization by inhibiting GABAergic neurons in the substantia nigra pars reticulata (SNr GABA ), and ultimately, selectively modulating the activity of ventral tegmental area dopamine (VTA DA ) cells projecting to the lateral shell of the nucleus accumbens (NAcLat). A major input to GPe PV cells is the indirect pathway of the dorsomedial striatum (DMS D 2 ), which receives DAergic innervation from collaterals of VTA DA →NAcLat cells, making this a closed-loop circuit. Cocaine likely facilitates reward and sensitization not directly through actions in the GPe, but rather in the upstream DMS, where the cocaine-induced elevation of DA triggers a depression in DMS D 2 cell activity. This cocaine-induced elevation in DA levels can be blocked by inhibition of GPe PV cells, closing the loop. Interestingly, the level of GPe PV cell activity prior to cocaine administration is correlated with the extent of reward and sensitization that animals experience in response to future administration of cocaine, indicating that GPe PV cell activity is a key predictor of future behavioral responses to cocaine. Single nucleus RNA-sequencing of GPe cells indicated that genes encoding voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability are downregulated in GPe PV cells following a single cocaine exposure, contributing to the elevation in GPe PV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPe PV cell excitability and also impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its potential as a therapeutic to counteract psychostimulant use disorder. Our findings illuminate the molecular and circuit mechanisms by which the GPe orchestrates brain-wide changes in response to cocaine that are required for reward, sensitization, and self-administration behaviors.
Collapse
|
8
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [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: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
9
|
Ramirez-Navarro A, Lima-Silveira L, Glazebrook PA, Dantzler HA, Kline DD, Kunze DL. Kv2 channels contribute to neuronal activity within the vagal afferent-nTS reflex arc. Am J Physiol Cell Physiol 2024; 326:C74-C88. [PMID: 37982174 PMCID: PMC11192486 DOI: 10.1152/ajpcell.00366.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
Diversity in the functional expression of ion channels contributes to the unique patterns of activity generated in visceral sensory A-type myelinated neurons versus C-type unmyelinated neurons in response to their natural stimuli. In the present study, Kv2 channels were identified as underlying a previously uncharacterized delayed rectifying potassium current expressed in both A- and C-type nodose ganglion neurons. Kv2.1 and 2.2 appear confined to the soma and initial segment of these sensory neurons; however, neither was identified in their central presynaptic terminals projecting onto relay neurons in the nucleus of the solitary tract (nTS). Kv2.1 and Kv2.2 were also not detected in the peripheral axons and sensory terminals in the aortic arch. Functionally, in nodose neuron somas, Kv2 currents exhibited frequency-dependent current inactivation and contributed to action potential repolarization in C-type neurons but not A-type neurons. Within the nTS, the block of Kv2 currents does not influence afferent presynaptic calcium influx or glutamate release in response to afferent activation, supporting our immunohistochemical observations. On the other hand, Kv2 channels contribute to membrane hyperpolarization and limit action potential discharge rate in second-order neurons. Together, these data demonstrate that Kv2 channels influence neuronal discharge within the vagal afferent-nTS circuit and indicate they may play a significant role in viscerosensory reflex function.NEW & NOTEWORTHY We demonstrate the expression and function of the voltage-gated delayed rectifier potassium channel Kv2 in vagal nodose neurons. Within sensory neurons, Kv2 channels limit the width of the broader C-type but not narrow A-type action potential. Within the nucleus of the solitary tract (nTS), the location of the vagal terminal field, Kv2 does not influence glutamate release. However, Kv2 limits the action potential discharge of nTS relay neurons. These data suggest a critical role for Kv2 in the vagal-nTS reflex arc.
Collapse
Affiliation(s)
- Angelina Ramirez-Navarro
- Rammelkamp Center for Education and Research, MetroHealth Medical Center Campus, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Ludmila Lima-Silveira
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Patricia A Glazebrook
- Rammelkamp Center for Education and Research, MetroHealth Medical Center Campus, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Heather A Dantzler
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - David D Kline
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Diana L Kunze
- Rammelkamp Center for Education and Research, MetroHealth Medical Center Campus, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
10
|
Huang Y, Ma D, Yang Z, Zhao Y, Guo J. Voltage-gated potassium channels KCNQs: Structures, mechanisms, and modulations. Biochem Biophys Res Commun 2023; 689:149218. [PMID: 37976835 DOI: 10.1016/j.bbrc.2023.149218] [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: 08/09/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
KCNQ (Kv7) channels are voltage-gated, phosphatidylinositol 4,5-bisphosphate- (PIP2-) modulated potassium channels that play essential roles in regulating the activity of neurons and cardiac myocytes. Hundreds of mutations in KCNQ channels are closely related to various cardiac and neurological disorders, such as long QT syndrome, epilepsy, and deafness, which makes KCNQ channels important drug targets. During the past several years, the application of single-particle cryo-electron microscopy (cryo-EM) technique in the structure determination of KCNQ channels has greatly advanced our understanding of their molecular mechanisms. In this review, we summarize the currently available structures of KCNQ channels, analyze their special voltage gating mechanism, and discuss their activation mechanisms by both the endogenous membrane lipid and the exogenous synthetic ligands. These structural studies of KCNQ channels will guide the development of drugs targeting KCNQ channels.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Cardiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Demin Ma
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenni Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yiwen Zhao
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050011, China
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Zahra A, Liu R, Wang J, Wu J. Identifying the mechanism of action of the Kv7 channel opener, retigabine in the treatment of epilepsy. Neurol Sci 2023; 44:3819-3825. [PMID: 37442907 DOI: 10.1007/s10072-023-06955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
Epilepsy is characterized by recurrent epileptic seizures caused by high levels of neuronal excitability in the brain. Voltage-sensitive K+ channels (Kv) of the Kv7 (KCNQ) family encoded by the KCNQ gene are involved in a wide range of cellular processes, i.e., KCNQ2 and KCNQ3 channels mediate M-currents to inhibit neuronal excitability and reduce transmitter release throughout the nervous system. Thus, as a positive allosteric modulator (or opener) of KCNQ channels, retigabine has been the only clinically approved anti-seizure medication that acts on the KCNQ channels. This review discusses the biochemical mechanisms about how retigabine acts on Kv7 channels, significance in neuronal pathophysiology, preclinical efficacy, and clinical stage of development. Additional efforts are being made to emphasize the possible benefits and drawbacks of retigabine compared to currently available medications for treatment-resistant epilepsy.
Collapse
Affiliation(s)
- Aqeela Zahra
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 122 Loushi Rd, Wuhan, 430070, China
- Department of Zoology, University of Sialkot, Sialkot, 51310, Pakistan
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
- National Clinical Research Center for Neurological Disease, Beijing, 100070, China
| | - Jingjing Wang
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 122 Loushi Rd, Wuhan, 430070, China
| | - Jianping Wu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 122 Loushi Rd, Wuhan, 430070, China.
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
- National Clinical Research Center for Neurological Disease, Beijing, 100070, China.
| |
Collapse
|
12
|
Ma D, Zheng Y, Li X, Zhou X, Yang Z, Zhang Y, Wang L, Zhang W, Fang J, Zhao G, Hou P, Nan F, Yang W, Su N, Gao Z, Guo J. Ligand activation mechanisms of human KCNQ2 channel. Nat Commun 2023; 14:6632. [PMID: 37857637 PMCID: PMC10587151 DOI: 10.1038/s41467-023-42416-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
The human voltage-gated potassium channel KCNQ2/KCNQ3 carries the neuronal M-current, which helps to stabilize the membrane potential. KCNQ2 can be activated by analgesics and antiepileptic drugs but their activation mechanisms remain unclear. Here we report cryo-electron microscopy (cryo-EM) structures of human KCNQ2-CaM in complex with three activators, namely the antiepileptic drug cannabidiol (CBD), the lipid phosphatidylinositol 4,5-bisphosphate (PIP2), and HN37 (pynegabine), an antiepileptic drug in the clinical trial, in an either closed or open conformation. The activator-bound structures, along with electrophysiology analyses, reveal the binding modes of two CBD, one PIP2, and two HN37 molecules in each KCNQ2 subunit, and elucidate their activation mechanisms on the KCNQ2 channel. These structures may guide the development of antiepileptic drugs and analgesics that target KCNQ2.
Collapse
Affiliation(s)
- Demin Ma
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Yueming Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Xiaoxiao Li
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Xiaoyu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Zhenni Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Yan Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Long Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenbo Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jiajia Fang
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Guohua Zhao
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Panpan Hou
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Fajun Nan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Nannan Su
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China.
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang, 311121, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
- State Key Laboratory of Plant Physiology and Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
13
|
Estrous Cycle Mediates Midbrain Neuron Excitability Altering Social Behavior upon Stress. J Neurosci 2023; 43:736-748. [PMID: 36549906 PMCID: PMC9899085 DOI: 10.1523/jneurosci.1504-22.2022] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/18/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
The estrous cycle is a potent modulator of neuron physiology. In rodents, in vivo ventral tegmental area (VTA) dopamine (DA) activity has been shown to fluctuate across the estrous cycle. Although the behavioral effect of fluctuating sex steroids on the reward circuit is well studied in response to drugs of abuse, few studies have focused on the molecular adaptations in the context of stress and motivated social behaviors. We hypothesized that estradiol fluctuations across the estrous cycle acts on the dopaminergic activity of the VTA to alter excitability and stress response. We used whole-cell slice electrophysiology of VTA DA neurons in naturally cycling, adult female C57BL/6J mice to characterize the effects of the estrous cycle and the role of 17β-estradiol on neuronal activity. We show that the estrous phase alters the effect of 17β-estradiol on excitability in the VTA. Behaviorally, the estrous phase during a series of acute variable social stressors modulates subsequent reward-related behaviors. Pharmacological inhibition of estrogen receptors in the VTA before stress during diestrus mimics the stress susceptibility found during estrus, whereas increased potassium channel activity in the VTA before stress reverses stress susceptibility found during estrus as assessed by social interaction behavior. This study identifies one possible potassium channel mechanism underlying the increased DA activity during estrus and reveals estrogen-dependent changes in neuronal function. Our findings demonstrate that the estrous cycle and estrogen signaling changes the physiology of DA neurons resulting in behavioral differences when the reward circuit is challenged with stress.SIGNIFICANCE STATEMENT The activity of the ventral tegmental area encodes signals of stress and reward. Dopaminergic activity has been found to be regulated by both local synaptic inputs as well as inputs from other brain regions. Here, we provide evidence that cycling sex steroids also play a role in modulating stress sensitivity of dopaminergic reward behavior. Specifically, we reveal a correlation of ionic activity with estrous phase, which influences the behavioral response to stress. These findings shed new light on how estrous cycle may influence dopaminergic activity primarily during times of stress perturbation.
Collapse
|
14
|
Jeong DJ, Kim KW, Suh BC. Dual regulation of Kv7.2/7.3 channels by long-chain n-alcohols. J Gen Physiol 2022; 155:213769. [PMID: 36534082 PMCID: PMC9767652 DOI: 10.1085/jgp.202213191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/31/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Normal alcohols (n-alcohols) can induce anesthetic effects by acting on neuronal ion channels. Recent studies have revealed the effects of n-alcohols on various ion channels; however, the underlying molecular mechanisms remain unclear. Here, we provide evidence that long-chain n-alcohols have dual effects on Kv7.2/7.3 channels, resulting in channel activation as the net effect. Using heterologous expression systems, we found that n-alcohols could differentially regulate the Kv7.2/7.3 channel depending on their chain length. Treatment with short-chain ethanol and propanol diminished Kv7.2/7.3 currents, whereas treatment with long-chain hexanol and octanol enhanced the currents. However, the long-chain alcohols failed to potentiate Kv7.2 currents pre-activated by retigabine. Instead, they inhibited the currents, similar to short-chain ethanol. The stimulatory effect of the long-chain n-alcohols was also converted into an inhibitory one in the mutant Kv7.2(W236L) channels, while the inhibitory effect of ethanol did not differ between wild-type Kv7.2 and mutant Kv7.2(W236L). The inhibition of currents by n-alcohols was also seen in Kv7.1 channel which does not have the tryptophan (W) residue in S5. These findings suggest that long-chain n-alcohols exhibit dual effects through independent working sites on the Kv7.2 channel. Finally, we confirmed that the hydroxyl group with a negative electrostatic potential surface is essential for the dual actions of n-alcohol. Together, our data suggest that long-chain n-alcohols regulate Kv7.2/7.3 channels by interacting with both stimulatory and inhibitory sites and that their stimulatory action depends on the conserved tryptophan 236 residue in S5 and could be important for triggering their anesthetic effects.
Collapse
Affiliation(s)
- Da-Jeong Jeong
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Kwon-Woo Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea,Correspondence to Byung-Chang Suh:
| |
Collapse
|
15
|
Paclitaxel Inhibits KCNQ Channels in Primary Sensory Neurons to Initiate the Development of Painful Peripheral Neuropathy. Cells 2022; 11:cells11244067. [PMID: 36552832 PMCID: PMC9776748 DOI: 10.3390/cells11244067] [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: 11/29/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer patients undergoing paclitaxel infusion usually experience peripheral nerve degeneration and serious neuropathic pain termed paclitaxel-induced peripheral neuropathy (PIPN). However, alterations in the dose or treatment schedule for paclitaxel do not eliminate PIPN, and no therapies are available for PIPN, despite numerous studies to uncover the mechanisms underlying the development/maintenance of this condition. Therefore, we aimed to uncover a novel mechanism underlying the pathogenesis of PIPN. Clinical studies suggest that acute over excitation of primary sensory neurons is linked to the pathogenesis of PIPN. We found that paclitaxel-induced acute hyperexcitability of primary sensory neurons results from the paclitaxel-induced inhibition of KCNQ potassium channels (mainly KCNQ2), found abundantly in sensory neurons and axons. We found that repeated application of XE-991, a specific KCNQ channel blocker, induced PIPN-like alterations in rats, including mechanical hypersensitivity and degeneration of peripheral nerves, as detected by both morphological and behavioral assays. In contrast, genetic deletion of KCNQ2 from peripheral sensory neurons in mice significantly attenuated the development of paclitaxel-induced peripheral sensory fiber degeneration and chronic pain. These findings may lead to a better understanding of the causes of PIPN and provide an impetus for developing new classes of KCNQ activators for its therapeutic treatment.
Collapse
|
16
|
Galán-Vidal J, Socuéllamos PG, Baena-Nuevo M, Contreras L, González T, Pérez-Poyato MS, Valenzuela C, González-Lamuño D, Gandarillas A. A novel loss-of-function mutation of the voltage-gated potassium channel Kv10.2 involved in epilepsy and autism. Orphanet J Rare Dis 2022; 17:345. [PMID: 36068614 PMCID: PMC9446776 DOI: 10.1186/s13023-022-02499-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Novel developmental mutations associated with disease are a continuous challenge in medicine. Clinical consequences caused by these mutations include neuron and cognitive alterations that can lead to epilepsy or autism spectrum disorders. Often, it is difficult to identify the physiological defects and the appropriate treatments. Results We have isolated and cultured primary cells from the skin of a patient with combined epilepsy and autism syndrome. A mutation in the potassium channel protein Kv10.2 was identified. We have characterised the alteration of the mutant channel and found that it causes loss of function (LOF). Primary cells from the skin displayed a very striking growth defect and increased differentiation. In vitro treatment with various carbonic anhydrase inhibitors with various degrees of specificity for potassium channels, (Brinzolamide, Acetazolamide, Retigabine) restored the activation capacity of the mutated channel. Interestingly, the drugs also recovered in vitro the expansion capacity of the mutated skin cells. Furthermore, treatment with Acetazolamide clearly improved the patient regarding epilepsy and cognitive skills. When the treatment was temporarily halted the syndrome worsened again. Conclusions By in vitro studying primary cells from the patient and the activation capacity of the mutated protein, we could first, find a readout for the cellular defects and second, test pharmaceutical treatments that proved to be beneficial. The results show the involvement of a novel LOF mutation of a Potassium channel in autism syndrome with epilepsy and the great potential of in vitro cultures of primary cells in personalised medicine of rare diseases.
Collapse
Affiliation(s)
- Jesús Galán-Vidal
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Paula G Socuéllamos
- Instituto de Investigaciones Biomédicas Alberto Sols, IIBM, CSIC-UAM, Madrid, Spain.,Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - María Baena-Nuevo
- Instituto de Investigaciones Biomédicas Alberto Sols, IIBM, CSIC-UAM, Madrid, Spain.,Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lizbeth Contreras
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Teresa González
- Instituto de Investigaciones Biomédicas Alberto Sols, IIBM, CSIC-UAM, Madrid, Spain.,Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - María S Pérez-Poyato
- Neuropediatric, University Hospital Marqués de Valdecilla, 39008, Santander, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols, IIBM, CSIC-UAM, Madrid, Spain. .,Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Domingo González-Lamuño
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain. .,Paediatric Department, University of Cantabria University, Marqués de Valdecilla Hospital, 39008, Santander, Spain.
| | - Alberto Gandarillas
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain. .,INSERM, Occitanie Méditerranée, 34394, Montpellier, France.
| |
Collapse
|
17
|
Jing J, Dunbar C, Sonesra A, Chavez A, Park S, Yang R, Soh H, Lee M, Tzingounis AV, Cooper EC, Jiang X, Maheshwari A. Removal of KCNQ2 from parvalbumin-expressing interneurons improves anti-seizure efficacy of retigabine. Exp Neurol 2022; 355:114141. [PMID: 35691372 PMCID: PMC9899633 DOI: 10.1016/j.expneurol.2022.114141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023]
Abstract
Anti-seizure drug (ASD) targets are widely expressed in both excitatory and inhibitory neurons. It remains unknown if the action of an ASD upon inhibitory neurons could counteract its beneficial effects on excitatory neurons (or vice versa), thereby reducing the efficacy of the ASD. Here, we examine whether the efficacy of the ASD retigabine (RTG) is altered after removal of the Kv7 potassium channel subunit KCNQ2, one of its drug targets, from parvalbumin-expressing interneurons (PV-INs). Parvalbumin-Cre (PV-Cre) mice were crossed with Kcnq2-floxed (Kcnq2fl/fl) mice to conditionally delete Kcnq2 from PV-INs. In these conditional knockout mice (cKO, PV-Kcnq2fl/fl), RTG (10 mg/kg, i.p.) significantly delayed the onset of either picrotoxin (PTX, 10 mg/kg, i.p)- or kainic acid (KA, 30 mg/kg, i.p.)-induced convulsive seizures compared to vehicle, while RTG was not effective in wild-type littermates (WT). Immunostaining for KCNQ2 and KCNQ3 revealed that both subunits were enriched at axon initial segments (AISs) of hippocampal CA1 PV-INs, and their specific expression was selectively abolished in cKO mice. Accordingly, the M-currents recorded from CA1 PV-INs and their sensitivity to RTG were significantly reduced in cKO mice. While the ability of RTG to suppress CA1 excitatory neurons in hippocampal slices was unchanged in cKO mice, its suppressive effect on the spike activity of CA1 PV-INs was significantly reduced compared with WT mice. In addition, the RTG-induced suppression on intrinsic membrane excitability of PV-INs in WT mice was significantly reduced in cKO mice. These findings suggest that preventing RTG from suppressing PV-INs improves its anticonvulsant effect.
Collapse
Affiliation(s)
- Junzhan Jing
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, United States of America
| | - Corrinne Dunbar
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Alina Sonesra
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Ana Chavez
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Suhyeorn Park
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Ryan Yang
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Heun Soh
- Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Rd, Storrs, CT 06269, United States of America
| | - Maxwell Lee
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Anastasios V Tzingounis
- Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Rd, Storrs, CT 06269, United States of America
| | - Edward C Cooper
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, United States of America.
| | - Atul Maheshwari
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| |
Collapse
|
18
|
Yang ND, Kanyo R, Zhao L, Li J, Kang PW, Dou AK, White KM, Shi J, Nerbonne JM, Kurata HT, Cui J. Electro-mechanical coupling of KCNQ channels is a target of epilepsy-associated mutations and retigabine. SCIENCE ADVANCES 2022; 8:eabo3625. [PMID: 35857840 PMCID: PMC9299555 DOI: 10.1126/sciadv.abo3625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
KCNQ2 and KCNQ3 form the M-channels that are important in regulating neuronal excitability. Inherited mutations that alter voltage-dependent gating of M-channels are associated with neonatal epilepsy. In the homolog KCNQ1 channel, two steps of voltage sensor activation lead to two functionally distinct open states, the intermediate-open (IO) and activated-open (AO), which define the gating, physiological, and pharmacological properties of KCNQ1. However, whether the M-channel shares the same mechanism is unclear. Here, we show that KCNQ2 and KCNQ3 feature only a single conductive AO state but with a conserved mechanism for the electro-mechanical (E-M) coupling between voltage sensor activation and pore opening. We identified some epilepsy-linked mutations in KCNQ2 and KCNQ3 that disrupt E-M coupling. The antiepileptic drug retigabine rescued KCNQ3 currents that were abolished by a mutation disrupting E-M coupling, suggesting that modulating the E-M coupling in KCNQ channels presents a potential strategy for antiepileptic therapy.
Collapse
Affiliation(s)
- Nien-Du Yang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Richard Kanyo
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Lu Zhao
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Jingru Li
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Po Wei Kang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Alex Kelly Dou
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Kelli McFarland White
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| | - Jeanne M. Nerbonne
- Departments of Developmental Biology and Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harley T. Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, and Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO 63130, USA
| |
Collapse
|
19
|
Development of KVO treatment strategies for chronic pain in a rat model of Gulf War Illness. Toxicol Appl Pharmacol 2022; 434:115821. [PMID: 34896435 DOI: 10.1016/j.taap.2021.115821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 12/19/2022]
Abstract
We examined whether combinations of Kv7 channel openers could be effective modifiers of deep tissue nociceptor activity; and whether such combinations could then be optimized for use as safe analgesics for pain-like signs that developed in a rat model of GWI (Gulf War Illness) pain. Voltage clamp experiments were performed on subclassified nociceptors isolated from rat DRG (dorsal root ganglion). A stepped voltage protocol was applied (-55 to -40 mV; Vh = -60 mV; 1500 ms) and Kv7 evoked currents were subsequently isolated by linopirdine subtraction. Directly activated and voltage activated K+ currents were characterized in the presence and absence of Retigabine (5-100 μM) and/or Diclofenac (50-140 μM). Retigabine produced substantial voltage dependent effects and a maximal sustained current of 1.14 pA/pF ± 0.15 (ED50: 62.7 ± 3.18 μM). Diclofenac produced weak voltage dependent effects but a similar maximum sustained current of 1.01 ± 0.26 pA/pF (ED50: 93.2 ± 8.99 μM). Combinations of Retigabine and Diclofenac substantially amplified resting currents but had little effect on voltage dependence. Using a cholinergic challenge test (Oxotremorine, 10 μM) associated with our GWI rat model, combinations of Retigabine (5 uM) and Diclofenac (2.5, 20 and 50 μM) substantially reduced or totally abrogated action potential discharge to the cholinergic challenge. When combinations of Retigabine and Diclofenac were used to relieve pain-signs in our rat model of GWI, only those combinations associated with serious subacute side effects could relieve pain-like behaviors.
Collapse
|
20
|
Abbott GW, Redford KE, Yoshimura RF, Manville RW, Moreira L, Tran K, Arena G, Kookootsedes A, Lasky E, Gunnison E. KCNQ and KCNE Isoform-Dependent Pharmacology Rationalizes Native American Dual Use of Specific Plants as Both Analgesics and Gastrointestinal Therapeutics. Front Physiol 2021; 12:777057. [PMID: 34858215 PMCID: PMC8632246 DOI: 10.3389/fphys.2021.777057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Indigenous peoples of the Americas are proficient in botanical medicine. KCNQ family voltage-gated potassium (Kv) channels are sensitive to a variety of ligands, including plant metabolites. Here, we screened methanolic extracts prepared from 40 Californian coastal redwood forest plants for effects on Kv current and membrane potential in Xenopus oocytes heterologously expressing KCNQ2/3, which regulates excitability of neurons, including those that sense pain. Extracts from 9 of the 40 plant species increased KCNQ2/3 current at –60 mV by ≥threefold (maximally, 15-fold by Urtica dioica) and/or hyperpolarized membrane potential by ≥-3 mV (maximally, –11 mV by Arctostaphylos glandulosa). All nine plants have traditionally been used as both analgesics and gastrointestinal therapeutics. Of two extracts tested, both acted as KCNQ-dependent analgesics in mice. KCNQ2/3 activation at physiologically relevant, subthreshold membrane potentials by tannic acid, gallic acid and quercetin provided molecular correlates for analgesic action of several of the plants. While tannic acid also activated KCNQ1 and KCNQ1-KCNE1 at hyperpolarized, negative membrane potentials, it inhibited KCNQ1-KCNE3 at both negative and positive membrane potentials, mechanistically rationalizing historical use of tannic acid-containing plants as gastrointestinal therapeutics. KCNE dependence of KCNQ channel modulation by plant metabolites therefore provides a molecular mechanistic basis for Native American use of specific plants as both analgesics and gastrointestinal aids.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Kaitlyn E Redford
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Ryan F Yoshimura
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Luiz Moreira
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Kevin Tran
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Grey Arena
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, United States
| | | | - Emma Lasky
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, United States
| | - Elliot Gunnison
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, United States
| |
Collapse
|
21
|
Aiba I, Noebels JL. Kcnq2/Kv7.2 controls the threshold and bi-hemispheric symmetry of cortical spreading depolarization. Brain 2021; 144:2863-2878. [PMID: 33768249 PMCID: PMC8536937 DOI: 10.1093/brain/awab141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/22/2021] [Accepted: 03/20/2021] [Indexed: 12/03/2022] Open
Abstract
Spreading depolarization is a slowly propagating wave of massive cellular depolarization associated with acute brain injury and migraine aura. Genetic studies link depolarizing molecular defects in Ca2+ flux, Na+ current in interneurons, and glial Na+-K+ ATPase with spreading depolarization susceptibility, emphasizing the important roles of synaptic activity and extracellular ionic homeostasis in determining spreading depolarization threshold. In contrast, although gene mutations in voltage-gated potassium ion channels that shape intrinsic membrane excitability are frequently associated with epilepsy susceptibility, it is not known whether epileptogenic mutations that regulate membrane repolarization also modify spreading depolarization threshold and propagation. Here we report that the Kcnq2/Kv7.2 potassium channel subunit, frequently mutated in developmental epilepsy, is a spreading depolarization modulatory gene with significant control over the seizure-spreading depolarization transition threshold, bi-hemispheric cortical expression, and diurnal temporal susceptibility. Chronic DC-band cortical EEG recording from behaving conditional Kcnq2 deletion mice (Emx1cre/+::Kcnq2flox/flox) revealed spontaneous cortical seizures and spreading depolarization. In contrast to the related potassium channel deficient model, Kv1.1-KO mice, spontaneous cortical spreading depolarizations in Kcnq2 cKO mice are tightly coupled to the terminal phase of seizures, arise bilaterally, and are observed predominantly during the dark phase. Administration of the non-selective Kv7.2 inhibitor XE991 to Kv1.1-KO mice partly reproduced the Kcnq2 cKO-like spreading depolarization phenotype (tight seizure coupling and bilateral symmetry) in these mice, indicating that Kv7.2 currents can directly and actively modulate spreading depolarization properties. In vitro brain slice studies confirmed that Kcnq2/Kv7.2 depletion or pharmacological inhibition intrinsically lowers the cortical spreading depolarization threshold, whereas pharmacological Kv7.2 activators elevate the threshold to multiple depolarizing and hypometabolic spreading depolarization triggers. Together these results identify Kcnq2/Kv7.2 as a distinctive spreading depolarization regulatory gene, and point to spreading depolarization as a potentially significant pathophysiological component of KCNQ2-linked epileptic encephalopathy syndromes. Our results also implicate KCNQ2/Kv7.2 channel activation as a potential adjunctive therapeutic target to inhibit spreading depolarization incidence.
Collapse
Affiliation(s)
- Isamu Aiba
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey L Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Larsson JE, Karlsson U, Wu X, Liin SI. Combining endocannabinoids with retigabine for enhanced M-channel effect and improved KV7 subtype selectivity. J Gen Physiol 2021; 152:151732. [PMID: 32365171 PMCID: PMC7398146 DOI: 10.1085/jgp.202012576] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022] Open
Abstract
Retigabine is unique among anticonvulsant drugs by targeting the neuronal M-channel, which is composed of KV7.2/KV7.3 and contributes to the negative neuronal resting membrane potential. Unfortunately, retigabine causes adverse effects, which limits its clinical use. Adverse effects may be reduced by developing M-channel activators with improved KV7 subtype selectivity. The aim of this study was to evaluate the prospect of endocannabinoids as M-channel activators, either in isolation or combined with retigabine. Human KV7 channels were expressed in Xenopus laevis oocytes. The effect of extracellular application of compounds with different properties was studied using two-electrode voltage clamp electrophysiology. Site-directed mutagenesis was used to construct channels with mutated residues to aid in the mechanistic understanding of these effects. We find that arachidonoyl-L-serine (ARA-S), a weak endocannabinoid, potently activates the human M-channel expressed in Xenopus oocytes. Importantly, we show that ARA-S activates the M-channel via a different mechanism and displays a different KV7 subtype selectivity compared with retigabine. We demonstrate that coapplication of ARA-S and retigabine at low concentrations retains the effect on the M-channel while limiting effects on other KV7 subtypes. Our findings suggest that improved KV7 subtype selectivity of M-channel activators can be achieved through strategically combining compounds with different subtype selectivity.
Collapse
Affiliation(s)
- Johan E Larsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Urban Karlsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xiongyu Wu
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Sara I Liin
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Naffaa MM, Al-Ewaidat OA. Ligand modulation of KCNQ-encoded (K V7) potassium channels in the heart and nervous system. Eur J Pharmacol 2021; 906:174278. [PMID: 34174270 DOI: 10.1016/j.ejphar.2021.174278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA.
| | - Ola A Al-Ewaidat
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
24
|
Ottosson NE, Silverå Ejneby M, Wu X, Estrada-Mondragón A, Nilsson M, Karlsson U, Schupp M, Rognant S, Jepps TA, Konradsson P, Elinder F. Synthetic resin acid derivatives selectively open the hK V 7.2/7.3 channel and prevent epileptic seizures. Epilepsia 2021; 62:1744-1758. [PMID: 34085706 DOI: 10.1111/epi.16932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE About one third of all patients with epilepsy have pharmacoresistant seizures. Thus there is a need for better pharmacological treatments. The human voltage-gated potassium (hKV ) channel hKV 7.2/7.3 is a validated antiseizure target for compounds that activate this channel. In a previous study we have shown that resin acid derivatives can activate the hKV 7.2/7.3 channel. In this study we investigated if these channel activators have the potential to be developed into a new type of antiseizure drug. Thus we examined their structure-activity relationships and the site of action on the hKV 7.2/7.3 channel, if they have unwanted cardiac and cardiovascular effects, and their potential antiseizure effect. METHODS Ion channels were expressed in Xenopus oocytes or mammalian cell lines and explored with two-electrode voltage-clamp or automated patch-clamp techniques. Unwanted vascular side effects were investigated with isometric tension recordings. Antiseizure activity was studied in an electrophysiological zebrafish-larvae model. RESULTS Fourteen resin acid derivatives were tested on hKV 7.2/7.3. The most efficient channel activators were halogenated and had a permanently negatively charged sulfonyl group. The compounds did not bind to the sites of other hKV 7.2/7.3 channel activators, retigabine, or ICA-069673. Instead, they interacted with the most extracellular gating charge of the S4 voltage-sensing helix, and the effects are consistent with an electrostatic mechanism. The compounds altered the voltage dependence of hKV 7.4, but in contrast to retigabine, there were no effects on the maximum conductance. Consistent with these data, the compounds had less smooth muscle-relaxing effect than retigabine. The compounds had almost no effect on the voltage dependence of hKV 11.1, hNaV 1.5, or hCaV 1.2, or on the amplitude of hKV 11.1. Finally, several resin acid derivatives had clear antiseizure effects in a zebrafish-larvae model. SIGNIFICANCE The described resin acid derivatives hold promise for new antiseizure medications, with reduced risk for adverse effects compared with retigabine.
Collapse
Affiliation(s)
- Nina E Ottosson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Malin Silverå Ejneby
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xiongyu Wu
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | | | - Michelle Nilsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Urban Karlsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Salomé Rognant
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Andrew Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Konradsson
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Fredrik Elinder
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
25
|
Dwivedi D, Bhalla US. Physiology and Therapeutic Potential of SK, H, and M Medium AfterHyperPolarization Ion Channels. Front Mol Neurosci 2021; 14:658435. [PMID: 34149352 PMCID: PMC8209339 DOI: 10.3389/fnmol.2021.658435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
SK, HCN, and M channels are medium afterhyperpolarization (mAHP)-mediating ion channels. The three channels co-express in various brain regions, and their collective action strongly influences cellular excitability. However, significant diversity exists in the expression of channel isoforms in distinct brain regions and various subcellular compartments, which contributes to an equally diverse set of specific neuronal functions. The current review emphasizes the collective behavior of the three classes of mAHP channels and discusses how these channels function together although they play specialized roles. We discuss the biophysical properties of these channels, signaling pathways that influence the activity of the three mAHP channels, various chemical modulators that alter channel activity and their therapeutic potential in treating various neurological anomalies. Additionally, we discuss the role of mAHP channels in the pathophysiology of various neurological diseases and how their modulation can alleviate some of the symptoms.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Stanley Center at the Broad, Cambridge, MA, United States
| | - Upinder S Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India
| |
Collapse
|
26
|
KCNQ Channels in the Mesolimbic Reward Circuit Regulate Nociception in Chronic Pain in Mice. Neurosci Bull 2021; 37:597-610. [PMID: 33900570 PMCID: PMC8099961 DOI: 10.1007/s12264-021-00668-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022] Open
Abstract
Mesocorticolimbic dopaminergic (DA) neurons have been implicated in regulating nociception in chronic pain, yet the mechanisms are barely understood. Here, we found that chronic constructive injury (CCI) in mice increased the firing activity and decreased the KCNQ channel-mediated M-currents in ventral tegmental area (VTA) DA neurons projecting to the nucleus accumbens (NAc). Chemogenetic inhibition of the VTA-to-NAc DA neurons alleviated CCI-induced thermal nociception. Opposite changes in the firing activity and M-currents were recorded in VTA DA neurons projecting to the medial prefrontal cortex (mPFC) but did not affect nociception. In addition, intra-VTA injection of retigabine, a KCNQ opener, while reversing the changes of the VTA-to-NAc DA neurons, alleviated CCI-induced nociception, and this was abolished by injecting exogenous BDNF into the NAc. Taken together, these findings highlight a vital role of KCNQ channel-mediated modulation of mesolimbic DA activity in regulating thermal nociception in the chronic pain state.
Collapse
|
27
|
Löscher W, Sills GJ, White HS. The ups and downs of alkyl-carbamates in epilepsy therapy: How does cenobamate differ? Epilepsia 2021; 62:596-614. [PMID: 33580520 DOI: 10.1111/epi.16832] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Since 1955, several alkyl-carbamates have been developed for the treatment of anxiety and epilepsy, including meprobamate, flupirtine, felbamate, retigabine, carisbamate, and cenobamate. They have each enjoyed varying levels of success as antiseizure drugs; however, they have all been plagued by the emergence of serious and sometimes life-threatening adverse events. In this review, we compare and contrast their predominant molecular mechanisms of action, their antiseizure profile, and where possible, their clinical efficacy. The preclinical, clinical, and mechanistic profile of the prototypical γ-aminobutyric acidergic (GABAergic) modulator phenobarbital is included for comparison. Like phenobarbital, all of the clinically approved alkyl-carbamates share an ability to enhance inhibitory neurotransmission through modulation of the GABAA receptor, although the specific mechanism of interaction differs among the different drugs discussed. In addition, several alkyl-carbamates have been shown to interact with voltage-gated ion channels. Flupirtine and retigabine share an ability to activate K+ currents mediated by KCNQ (Kv7) K+ channels, and felbamate, carisbamate, and cenobamate have been shown to block Na+ channels. In contrast to other alkyl-carbamates, cenobamate seems to be unique in its ability to preferentially attenuate the persistent rather than transient Na+ current. Results from recent randomized controlled clinical trials with cenobamate suggest that this newest antiseizure alkyl-carbamate possesses a degree of efficacy not witnessed since felbamate was approved in 1993. Given that ceno-bamate's mechanistic profile is unique among the alkyl-carbamates, it is not clear whether this impressive efficacy reflects an as yet undescribed mechanism of action or whether it possesses a unique synergy between its actions at the GABAA receptor and on persistent Na+ currents. The high efficacy of cenobamate is, however, tempered by the risk of serious rash and low tolerability at higher doses, meaning that further safety studies and clinical experience are needed to determine the true clinical value of cenobamate.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Graeme J Sills
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
28
|
Li X, Zhang Q, Guo P, Fu J, Mei L, Lv D, Wang J, Lai D, Ye S, Yang H, Guo J. Molecular basis for ligand activation of the human KCNQ2 channel. Cell Res 2021; 31:52-61. [PMID: 32884139 PMCID: PMC7852908 DOI: 10.1038/s41422-020-00410-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/14/2020] [Indexed: 01/10/2023] Open
Abstract
The voltage-gated potassium channel KCNQ2 is responsible for M-current in neurons and is an important drug target to treat epilepsy, pain and several other diseases related to neuronal hyper-excitability. A list of synthetic compounds have been developed to directly activate KCNQ2, yet our knowledge of their activation mechanism is limited, due to lack of high-resolution structures. Here, we report cryo-electron microscopy (cryo-EM) structures of the human KCNQ2 determined in apo state and in complex with two activators, ztz240 or retigabine, which activate KCNQ2 through different mechanisms. The activator-bound structures, along with electrophysiology analysis, reveal that ztz240 binds at the voltage-sensing domain and directly stabilizes it at the activated state, whereas retigabine binds at the pore domain and activates the channel by an allosteric modulation. By accurately defining ligand-binding sites, these KCNQ2 structures not only reveal different ligand recognition and activation mechanisms, but also provide a structural basis for drug optimization and design.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Peipei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Jie Fu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Lianghe Mei
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 108 Yuxin Road, Suzhou, Jiangsu, 215123, China
| | - Dashuai Lv
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jiangqin Wang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Dongwu Lai
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Sheng Ye
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China.
| | - Jiangtao Guo
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
29
|
Liu Y, Bian X, Wang K. Pharmacological Activation of Neuronal Voltage-Gated Kv7/KCNQ/M-Channels for Potential Therapy of Epilepsy and Pain. Handb Exp Pharmacol 2021; 267:231-251. [PMID: 33837465 DOI: 10.1007/164_2021_458] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Native M-current is a low-threshold, slowly activating potassium current that exerts an inhibitory control over neuronal excitability. The M-channel is primarily co-assembled by heterotetrameric Kv7.2/KCNQ2 and Kv7.3/KCNQ3 subunits that are specifically expressed in the brain and peripheral nociceptive and visceral sensory neurons in the spinal cord. Reduction of M-channel function leads to neuronal hyperexcitability that defines the fundamental mechanism of neurological disorders such as epilepsy and pain, indicating that pharmacological activation of Kv7/KCNQ/M-channels may serve the basis for the therapy. The well-known KCNQ opener retigabine (ezogabine or Potiga) was approved by FDA in 2011 as an anticonvulsant used for an adjunctive treatment of partial epilepsies. Unfortunately, retigabine was discontinued in 2017 due to its side effects of blue-colored appearance of the skin and eyes after prolonged intake. In addition, flupirtine, a structural derivative of retigabine and a centrally acting non-opioid analgesic, was also withdrawn in 2018 for liver toxicity. Fortunately, these side effects are compound-structures related and can be avoided. Thus, further identification and development of novel potent and selective Kv7 channel openers may lead to an effective therapy with improved safety window for anti-epilepsy and anti-nociception.
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Xiling Bian
- Department of Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - KeWei Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China. .,Institute of Innovative Drugs Qingdao University, Qingdao, China.
| |
Collapse
|
30
|
Wu X, Larsson HP. Insights into Cardiac IKs (KCNQ1/KCNE1) Channels Regulation. Int J Mol Sci 2020; 21:ijms21249440. [PMID: 33322401 PMCID: PMC7763278 DOI: 10.3390/ijms21249440] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
The delayed rectifier potassium IKs channel is an important regulator of the duration of the ventricular action potential. Hundreds of mutations in the genes (KCNQ1 and KCNE1) encoding the IKs channel cause long QT syndrome (LQTS). LQTS is a heart disorder that can lead to severe cardiac arrhythmias and sudden cardiac death. A better understanding of the IKs channel (here called the KCNQ1/KCNE1 channel) properties and activities is of great importance to find the causes of LQTS and thus potentially treat LQTS. The KCNQ1/KCNE1 channel belongs to the superfamily of voltage-gated potassium channels. The KCNQ1/KCNE1 channel consists of both the pore-forming subunit KCNQ1 and the modulatory subunit KCNE1. KCNE1 regulates the function of the KCNQ1 channel in several ways. This review aims to describe the current structural and functional knowledge about the cardiac KCNQ1/KCNE1 channel. In addition, we focus on the modulation of the KCNQ1/KCNE1 channel and its potential as a target therapeutic of LQTS.
Collapse
|
31
|
Tsai YM, Jones F, Mullen P, Porter KE, Steele D, Peers C, Gamper N. Vascular Kv7 channels control intracellular Ca 2+ dynamics in smooth muscle. Cell Calcium 2020; 92:102283. [PMID: 32950876 PMCID: PMC7695684 DOI: 10.1016/j.ceca.2020.102283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 01/23/2023]
Abstract
Voltage-gated Kv7 (or KCNQ) channels control activity of excitable cells, including vascular smooth muscle cells (VSMCs), by setting their resting membrane potential and controlling other excitability parameters. Excitation-contraction coupling in muscle cells is mediated by Ca2+ but until now, the exact role of Kv7 channels in cytosolic Ca2+ dynamics in VSMCs has not been fully elucidated. We utilised microfluorimetry to investigate the impact of Kv7 channel activity on intracellular Ca2+ levels and electrical activity of rat A7r5 VSMCs and primary human internal mammary artery (IMA) SMCs. Both, direct (XE991) and G protein coupled receptor mediated (vasopressin, AVP) Kv7 channel inhibition induced robust Ca2+ oscillations, which were significantly reduced in the presence of Kv7 channel activator, retigabine, L-type Ca2+ channel inhibitor, nifedipine, or T-type Ca2+ channel inhibitor, NNC 55-0396, in A7r5 cells. Membrane potential measured using FluoVolt exhibited a slow depolarisation followed by a burst of sharp spikes in response to XE991; spikes were temporally correlated with Ca2+ oscillations. Phospholipase C inhibitor (edelfosine) reduced AVP-induced, but not XE991-induced Ca2+ oscillations. AVP and XE991 induced a large increase of [Ca2+]i in human IMA, which was also attenuated with retigabine, nifedipine and NNC 55-0396. RT-PCR, immunohistochemistry and electrophysiology suggested that Kv7.5 was the predominant Kv7 subunit in both rat and human arterial SMCs; CACNA1C (Cav1.2; L-type) and CACNA1 G (Cav3.1; T-type) were the most abundant voltage-gated Ca2+ channel gene transcripts in both types of VSMCs. This study establishes Kv7 channels as key regulators of Ca2+ signalling in VSMCs with Kv7.5 playing a dominant role.
Collapse
Affiliation(s)
- Yuan-Ming Tsai
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom; Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defence Medical Centre, Taipei 11490, Taiwan.
| | - Frederick Jones
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Pierce Mullen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Karen E Porter
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Derek Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Chris Peers
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
32
|
Lee SH, Kang J, Ho A, Watanabe H, Bolshakov VY, Shen J. APP Family Regulates Neuronal Excitability and Synaptic Plasticity but Not Neuronal Survival. Neuron 2020; 108:676-690.e8. [PMID: 32891188 PMCID: PMC7704911 DOI: 10.1016/j.neuron.2020.08.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 01/03/2023]
Abstract
Amyloid precursor protein (APP) is associated with both familial and sporadic forms of Alzheimer's disease. Despite its importance, the role of APP family in neuronal function and survival remains unclear because of perinatal lethality exhibited by knockout mice lacking all three APP family members. Here we report that selective inactivation of APP family members in excitatory neurons of the postnatal forebrain results in neither cortical neurodegeneration nor increases in apoptosis and gliosis up to ∼2 years of age. However, hippocampal synaptic plasticity, learning, and memory are impaired in these mutant mice. Furthermore, hippocampal neurons lacking APP family exhibit hyperexcitability, as evidenced by increased neuronal spiking in response to depolarizing current injections, whereas blockade of Kv7 channels mimics and largely occludes the effects of APP family inactivation. These findings demonstrate that APP family is not required for neuronal survival and suggest that APP family may regulate neuronal excitability through Kv7 channels.
Collapse
Affiliation(s)
- Sang Hun Lee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jongkyun Kang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angela Ho
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hirotaka Watanabe
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vadim Y Bolshakov
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Jie Shen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Davis H, Herring N, Paterson DJ. Downregulation of M Current Is Coupled to Membrane Excitability in Sympathetic Neurons Before the Onset of Hypertension. Hypertension 2020; 76:1915-1923. [PMID: 33040619 PMCID: PMC8360673 DOI: 10.1161/hypertensionaha.120.15922] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Neurohumoral activation is an early hallmark of cardiovascular disease and contributes to the etiology of the pathophysiology. Stellectomy has reemerged as a positive therapeutic intervention to modify the progression of dysautonomia, although the biophysical properties underpinning abnormal activity of this ganglia are not fully understood in the initial stages of the disease. We investigated whether stellate ganglia neurons from prehypertensive SHRs (spontaneously hypertensive rats) are hyperactive and describe their electrophysiological phenotype guided by single-cell RNA sequencing, molecular biology, and perforated patch clamp to uncover the mechanism of abnormal excitability. We demonstrate the contribution of a plethora of ion channels, in particular inhibition of M current to stellate ganglia neuronal firing, and confirm the conservation of expression of key ion channel transcripts in human stellate ganglia. We show that hyperexcitability was curbed by M-current activators, nonselective sodium current blockers, or inhibition of Nav1.1-1.3, Nav1.6, or INaP. We conclude that reduced activity of M current contributes significantly to abnormal firing of stellate neurons, which, in part, contributes to the hyperexcitability from rats that have a predisposition to hypertension. Targeting these channels could provide a therapeutic opportunity to minimize the consequences of excessive sympathetic activation.
Collapse
Affiliation(s)
- Harvey Davis
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, Wellcome Trust OXION Initiative in Ion Channels and Disease (H.D., D.J.P.), University of Oxford, United Kingdom
| | - Neil Herring
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, United Kingdom (N.H.)
| | - David J Paterson
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, Wellcome Trust OXION Initiative in Ion Channels and Disease (H.D., D.J.P.), University of Oxford, United Kingdom
| |
Collapse
|
34
|
Redford KE, Abbott GW. The ubiquitous flavonoid quercetin is an atypical KCNQ potassium channel activator. Commun Biol 2020; 3:356. [PMID: 32641720 PMCID: PMC7343821 DOI: 10.1038/s42003-020-1089-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Many commonly consumed plants are used as folk medicines, often with unclear molecular mechanisms. Recent studies uncovered the ubiquitous and influential KCNQ family of voltage-gated potassium (Kv) channels as a therapeutic target for several medicinal plant compounds. Capers - immature flower buds of Capparis spinosa - have been consumed for food and medicinal purposes for millennia. Here, we show that caper extract hyperpolarizes cells expressing KCNQ1 or KCNQ2/3 Kv channels. Capers are the richest known natural source of quercetin, the most consumed dietary flavonoid. Quercetin potentiated KCNQ1/KCNE1, KCNQ2/3 and KCNQ4 currents but, unusually, not KCNQ5. Strikingly, quercetin augmented both activation and inactivation of KCNQ1, via a unique KCNQ activation mechanism involving sites atop the voltage sensor and in the pore. The findings uncover a novel potential molecular basis for therapeutic effects of quercetin-rich foods and a new chemical space for atypical modes of KCNQ channel modulation. Kaitlyn E. Redford and Geoffrey W. Abbott show that quercetin, a flavonoid highly expressed in capers, potentiates KCNQ currents to varying degrees depending on the subunit composition of the channel complex. By combining in silico docking, mutagenesis, and electrophysiology they show that this flavonoid can bind KCNQ channels atop the voltage sensor and within the pore module, highlighting an atypical mode of channel modulation.
Collapse
Affiliation(s)
- Kaitlyn E Redford
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
35
|
Villalba-Galea CA. Modulation of K V7 Channel Deactivation by PI(4,5)P 2. Front Pharmacol 2020; 11:895. [PMID: 32636742 PMCID: PMC7318307 DOI: 10.3389/fphar.2020.00895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/02/2020] [Indexed: 01/16/2023] Open
Abstract
The activity of KV7 channels critically contributes to the regulation of cellular electrical excitability in many cell types. In the central nervous system, the heteromeric KV7.2/KV7.3 channel is thought to be the chief molecular entity giving rise to M-currents. These K+-currents as so called because they are inhibited by the activation of Gq protein-coupled muscarinic receptors. In general, activation of Gq protein-coupled receptors (GqPCRs) decreases the concentration of the phosphoinositide PI(4,5)P2 which is required for KV7 channel activity. It has been recently reported that the deactivation rate of KV7.2/KV7.3 channels decreases as a function of activation. This suggests that the activated/open channel stabilizes as activation persists. This property has been regarded as evidence for the existence of modal behavior in the activity of these channels. In particular, it has been proposed that the heteromeric KV7.2/KV7.3 channel has at least two modes of activity that can be distinguished by both their deactivation kinetics and sensitivity to Retigabine. The current study was aimed at understanding the effect of PI(4,5)P2 depletion on the modal behavior of KV7.2/KV7.3 channels. Here, it was hypothesized that depleting the membrane of P(4,5)P2 would hamper the stabilization of the activated/open channel, resulting in higher rates of deactivation of the heteromeric KV7.2/KV7.3 channel. In addressing this question, it was found that the activity-dependent slowdown of the deactivation was not as prominent when channels were co-expressed with the chimeric phosphoinositide-phosphatase Ci-VS-TPIP or when cells were treated with the phosphoinositide kinase inhibitor Wortmannin. Further, it was observed that either of these approaches to deplete PI(4,5)P2 had a higher impact on the kinetic of deactivation following prolonged activation, while having little or no effect when activation was short-lived. Furthermore, it was observed that the action of either Ci-VS-TPIP or Wortmannin reduced the effect of Retigabine on the kinetics of deactivation, having a higher impact when activation was prolonged. These combined observations led to the conclusion that the deactivation kinetic of KV7.2/KV7.3 channels was sensitive to PI(4,5)P2 depletion in an activation-dependent manner, displaying a stronger effect on deactivation following prolonged activation.
Collapse
Affiliation(s)
- Carlos A. Villalba-Galea
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
| |
Collapse
|
36
|
Vigil FA, Carver CM, Shapiro MS. Pharmacological Manipulation of K v 7 Channels as a New Therapeutic Tool for Multiple Brain Disorders. Front Physiol 2020; 11:688. [PMID: 32636759 PMCID: PMC7317068 DOI: 10.3389/fphys.2020.00688] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
K v 7 ("M-type," KCNQ) K+ currents, play dominant roles in controlling neuronal excitability. They act as a "brake" against hyperexcitable states in the central and peripheral nervous systems. Pharmacological augmentation of M current has been developed for controlling epileptic seizures, although current pharmacological tools are uneven in practical usefulness. Lately, however, M-current "opener" compounds have been suggested to be efficacious in preventing brain damage after multiple types of insults/diseases, such as stroke, traumatic brain injury, drug addiction and mood disorders. In this review, we will discuss what is known to date on these efforts and identify gaps in our knowledge regarding the link between M current and therapeutic potential for these disorders. We will outline the preclinical experiments that are yet to be performed to demonstrate the likelihood of success of this approach in human trials. Finally, we also address multiple pharmacological tools available to manipulate different K v 7 subunits and the relevant evidence for translational application in the clinical use for disorders of the central nervous system and multiple types of brain insults. We feel there to be great potential for manipulation of K v 7 channels as a novel therapeutic mode of intervention in the clinic, and that the paucity of existing therapies obligates us to perform further research, so that patients can soon benefit from such therapeutic approaches.
Collapse
Affiliation(s)
- Fabio A Vigil
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Mark S Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
37
|
Gore A, Neufeld-Cohen A, Egoz I, Baranes S, Gez R, Grauer E, Chapman S, Lazar S. Efficacy of retigabine in ameliorating the brain insult following sarin exposure in the rat. Toxicol Appl Pharmacol 2020; 395:114963. [PMID: 32209366 DOI: 10.1016/j.taap.2020.114963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Sarin is an irreversible organophosphate cholinesterase inhibitor. Following toxic signs, an extensive long-term brain damage is often reported. Thus, we evaluated the efficacy of a novel anticonvulsant drug retigabine, a modulator of neuronal voltage gated K+ channels, as a neuroprotective agent following sarin exposure. METHODS Rats were exposed to 1 LD50 or 1.2 LD50 sarin and treated at onset of convulsions with retigabine (5 mg/kg, i.p.) alone or in combination with 5 mg/kg atropine and 7.5 mg/kg TMB-4 (TA) respectively. Brain biochemical and immunohistopathological analyses were processed 24 h and 1 week following 1 LD50 sarin exposure and at 4 weeks following exposure to 1.2 LD50 sarin. EEG activity in freely moving rats was also monitored by telemetry during the first week following exposure to 1.2 LD50 and behavior in the Open Field was evaluated 3 weeks post exposure. RESULTS Treatment with retigabine following 1 LD50 sarin exposure or in combination with TA following 1.2 LD50 exposure significantly reduced mortality rate compared to the non-treated groups. In both experiments, the retigabine treatment significantly reduced gliosis, astrocytosis and brain damage as measured by translocator protein (TSPO). Following sarin exposure the combined treatment (retigabine+ TA) significantly minimized epileptiform seizure activity. Finally, in the Open Field behavioral test the non-treated sarin group showed an increased mobility which was reversed by the combined treatment. CONCLUSIONS The M current modulator retigabine has been shown to be an effective adjunct therapy following OP induced convulsion, minimizing epileptiform seizure activity and attenuating the ensuing brain damage.
Collapse
Affiliation(s)
- Ariel Gore
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel.
| | - Adi Neufeld-Cohen
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Inbal Egoz
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Shlomi Baranes
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Rellie Gez
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Ettie Grauer
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Shira Chapman
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Shlomi Lazar
- Department. of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel.
| |
Collapse
|
38
|
Shi S, Li J, Sun F, Chen Y, Pang C, Geng Y, Qi J, Guo S, Wang X, Zhang H, Zhan Y, An H. Molecular Mechanisms and Structural Basis of Retigabine Analogues in Regulating KCNQ2 Channel. J Membr Biol 2020; 253:167-181. [DOI: 10.1007/s00232-020-00113-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/05/2020] [Indexed: 12/22/2022]
|
39
|
Manville RW, Papanikolaou M, Abbott GW. M-Channel Activation Contributes to the Anticonvulsant Action of the Ketone Body β-Hydroxybutyrate. J Pharmacol Exp Ther 2020; 372:148-156. [PMID: 31757819 PMCID: PMC6994816 DOI: 10.1124/jpet.119.263350] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/20/2019] [Indexed: 12/26/2022] Open
Abstract
Ketogenic diets are effective therapies for refractory epilepsy, yet the underlying mechanisms are incompletely understood. The anticonvulsant efficacy of ketogenic diets correlates positively to the serum concentration of β-hydroxybutyrate (BHB), the primary ketone body generated by ketosis. Voltage-gated potassium channels generated by KCNQ2-5 subunits, especially KCNQ2/3 heteromers, generate the M-current, a therapeutic target for synthetic anticonvulsants. Here, we report that BHB directly activates KCNQ2/3 channels (EC50 = 0.7 µM), via a highly conserved S5 tryptophan (W265) on KCNQ3. BHB was also acutely effective as an anticonvulsant in the pentylene tetrazole (PTZ) seizure assay in mice. Strikingly, coadministration of γ-amino-β-hydroxybutyric acid, a high-affinity KCNQ2/3 partial agonist that also acts via KCNQ3-W265, similarly reduced the efficacy of BHB in KCNQ2/3 channel activation in vitro and in the PTZ seizure assay in vivo. Our results uncover a novel, unexpected molecular basis for anticonvulsant effects of the major ketone body induced by ketosis. SIGNIFICANCE STATEMENT: Ketogenic diets are used to treat refractory epilepsy but the therapeutic mechanism is not fully understood. Here, we show that clinically relevant concentrations of β-hydroxybutyrate, the primary ketone body generated during ketogenesis, activates KCNQ2/3 potassium channels by binding to a specific site on KCNQ3, an effect known to reduce neuronal excitability. We provide evidence using a mouse chemoconvulsant model that KCNQ2/3 activation contributes to the antiepileptic action of β-hydroxybutyrate.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| | - Maria Papanikolaou
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| |
Collapse
|
40
|
Ostacolo C, Miceli F, Di Sarno V, Nappi P, Iraci N, Soldovieri MV, Ciaglia T, Ambrosino P, Vestuto V, Lauritano A, Musella S, Pepe G, Basilicata MG, Manfra M, Perinelli DR, Novellino E, Bertamino A, Gomez-Monterrey IM, Campiglia P, Taglialatela M. Synthesis and Pharmacological Characterization of Conformationally Restricted Retigabine Analogues as Novel Neuronal Kv7 Channel Activators. J Med Chem 2019; 63:163-185. [DOI: 10.1021/acs.jmedchem.9b00796] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Carmine Ostacolo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Francesco Miceli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, 80131 Naples, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Piera Nappi
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, 80131 Naples, Italy
| | - Nunzio Iraci
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Maria Virginia Soldovieri
- Department of Medicine and Health Science V. Tiberio, University of Molise, Via F. de Sanctis, 86100 Campobasso, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Paolo Ambrosino
- Department of Science and Technology (DST), University of Sannio, Via Port’Arsa 11, 82100 Benevento, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Anna Lauritano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, 80131 Naples, Italy
| | - Simona Musella
- European Biomedical Research Center (EBRIS), Via Salvatore de Renzi, 3, 84125 Salerno, Salerno, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | | | - Michele Manfra
- Department of Science, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Diego Romano Perinelli
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Macerata, Italy
| | - Ettore Novellino
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | | | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Maurizio Taglialatela
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
41
|
Metzger S, Dupont C, Voss AA, Rich MM. Central Role of Subthreshold Currents in Myotonia. Ann Neurol 2019; 87:175-183. [PMID: 31725924 DOI: 10.1002/ana.25646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 01/11/2023]
Abstract
It is generally thought that muscle excitability is almost exclusively controlled by currents responsible for generation of action potentials. We propose that smaller ion channel currents that contribute to setting the resting potential and to subthreshold fluctuations in membrane potential can also modulate excitability in important ways. These channels open at voltages more negative than the action potential threshold and are thus termed subthreshold currents. As subthreshold currents are orders of magnitude smaller than the currents responsible for the action potential, they are hard to identify and easily overlooked. Discovery of their importance in regulation of excitability opens new avenues for improved therapy for muscle channelopathies and diseases of the neuromuscular junction. ANN NEUROL 2020;87:175-183.
Collapse
Affiliation(s)
- Sabrina Metzger
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Chris Dupont
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Andrew A Voss
- Department of Biology, Wright State University, Dayton, OH
| | - Mark M Rich
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| |
Collapse
|
42
|
Corydon KK, Matchkov V, Fais R, Abramochkin D, Hedegaard ER, Comerma-Steffensen S, Simonsen U. Effect of ischemic preconditioning and a Kv7 channel blocker on cardiac ischemia-reperfusion injury in rats. Eur J Pharmacol 2019; 866:172820. [PMID: 31760069 DOI: 10.1016/j.ejphar.2019.172820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/30/2022]
Abstract
Recently, we found cardioprotective effects of ischemic preconditioning (IPC), and from a blocker of KCNQ voltage-gated K+ channels (KV7), XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone), in isolated rat hearts. The purpose of the present study was to investigate the cardiovascular effects of IPC and XE991 and whether they are cardioprotective in intact rats. In conscious rats, we measured the effect of the KV7 channel blocker XE991 on heart rate and blood pressure by use of telemetry. In anesthetized rats, cardiac ischemia was induced by occluding the left coronary artery, and the animals received IPC (2 × 5 min of occlusion), XE991, or a combination. After a 2 h reperfusion period, the hearts were excised, and the area at risk and infarct size were determined. In both anesthetized and conscious rats, XE991 increased blood pressure, and the highest dose (7.5 mg/kg) of XE991 also increased heart rate, and 44% of conscious rats died. XE991 induced marked changes in the electrocardiogram (e.g., increased PR interval and prolonged QTC interval) without changing cardiac action potentials. The infarct size to area at risk ratio was reduced from 53 ± 2% (n = 8) in the vehicle compared to 36 ± 3% in the IPC group (P < 0.05, n = 9). XE991 (0.75 mg/kg) treatment alone or on top of IPC failed to reduce myocardial infarct size. Similar to the effect in isolated hearts, locally applied IPC was cardioprotective in intact animals exposed to ischemia-reperfusion. Systemic administration of XE991 failed to protect the heart against ischemia-reperfusion injury suggesting effects on the autonomic nervous system counteracting the cardioprotection in intact animals.
Collapse
Affiliation(s)
- Krestine Kjeldsen Corydon
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Vladimir Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Rafael Fais
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Denis Abramochkin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia; Ural Federal University, Mira 19, Ekaterinburg, Russia; Department of Physiology, Russian National Research Medical University, Ostrovityanova 1, Moscow, Russia
| | - Elise Røge Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Biomedical Sciences/Animal Physiology, Veterinary Faculty, Central University of Venezuela, Maracay, Aragua, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark.
| |
Collapse
|
43
|
Manville RW, Abbott GW. In silico re-engineering of a neurotransmitter to activate KCNQ potassium channels in an isoform-specific manner. Commun Biol 2019; 2:401. [PMID: 31701029 PMCID: PMC6825221 DOI: 10.1038/s42003-019-0648-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated potassium (Kv) channel dysfunction causes a variety of inherited disorders, but developing small molecules that activate Kv channels has proven challenging. We recently discovered that the inhibitory neurotransmitter γ-aminobutyric acid (GABA) directly activates Kv channels KCNQ3 and KCNQ5. Here, finding that inhibitory neurotransmitter glycine does not activate KCNQs, we re-engineered it in silico to introduce predicted KCNQ-opening properties, screened by in silico docking, then validated the hits in vitro. Attaching a fluorophenyl ring to glycine optimized its electrostatic potential, converting it to a low-nM affinity KCNQ channel activator. Repositioning the phenyl ring fluorine and/or adding a methylsulfonyl group increased the efficacy of the re-engineered glycines and switched their target KCNQs. Combining KCNQ2- and KCNQ3-specific glycine derivatives synergistically potentiated KCNQ2/3 activation by exploiting heteromeric channel composition. Thus, in silico optimization and docking, combined with functional screening of only three compounds, facilitated re-engineering of glycine to develop several potent KCNQ activators.
Collapse
Affiliation(s)
- Rían W. Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA USA
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA USA
| |
Collapse
|
44
|
Manville RW, Abbott GW. Cilantro leaf harbors a potent potassium channel-activating anticonvulsant. FASEB J 2019; 33:11349-11363. [PMID: 31311306 PMCID: PMC6766653 DOI: 10.1096/fj.201900485r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/25/2019] [Indexed: 11/11/2022]
Abstract
Herbs have a long history of use as folk medicine anticonvulsants, yet the underlying mechanisms often remain unknown. Neuronal voltage-gated potassium channel subfamily Q (KCNQ) dysfunction can cause severe epileptic encephalopathies that are resistant to modern anticonvulsants. Here we report that cilantro (Coriandrum sativum), a widely used culinary herb that also exhibits antiepileptic and other therapeutic activities, is a highly potent KCNQ channel activator. Screening of cilantro leaf metabolites revealed that one, the long-chain fatty aldehyde (E)-2-dodecenal, activates multiple KCNQs, including the predominant neuronal isoform, KCNQ2/KCNQ3 [half maximal effective concentration (EC50), 60 ± 20 nM], and the predominant cardiac isoform, KCNQ1 in complexes with the type I transmembrane ancillary subunit (KCNE1) (EC50, 260 ± 100 nM). (E)-2-dodecenal also recapitulated the anticonvulsant action of cilantro, delaying pentylene tetrazole-induced seizures. In silico docking and mutagenesis studies identified the (E)-2-dodecenal binding site, juxtaposed between residues on the KCNQ S5 transmembrane segment and S4-5 linker. The results provide a molecular basis for the therapeutic actions of cilantro and indicate that this ubiquitous culinary herb is surprisingly influential upon clinically important KCNQ channels.-Manville, R. W., Abbott, G. W. Cilantro leaf harbors a potent potassium channel-activating anticonvulsant.
Collapse
Affiliation(s)
- Rían W. Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California–Irvine, Irvine, California, USA
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California–Irvine, Irvine, California, USA
| |
Collapse
|
45
|
Lyman KA, Chetkovich DM. Oligodendrocyte K ir4.1 Channels Clear Out Congested K .. Epilepsy Curr 2019; 19:339-340. [PMID: 31409147 PMCID: PMC6864574 DOI: 10.1177/1535759719868185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Oligodendrocytes Control Potassium Accumulation in White Matter and Seizure Susceptibility. Larson VA, Mironova Y, Vanderpool KG, Waisman A, Rash JE, Agarwal A, Bergles DE. Elife. 2018 Mar 29;7. pii: e34829. doi:10.7554/eLife.34829. The inwardly rectifying K+ channel Kir4.1 is broadly expressed by central nervous system glia and deficits in Kir4.1 lead to seizures and myelin vacuolization. However, the role of oligodendrocyte Kir4.1 channels in controlling myelination and K+ clearance in white matter has not been defined. Here, we show that selective deletion of Kir4.1 from oligodendrocyte progenitors or mature oligodendrocytes did not impair their development or disrupt the structure of myelin. However, mice lacking oligodendrocyte Kir4.1 channels exhibited profound functional impairments, including slower clearance of extracellular K+ and delayed recovery of axons from repetitive stimulation in white matter, as well as spontaneous seizures, a lower seizure threshold, and activity-dependent motor deficits. These results indicate that Kir4.1 channels in oligodendrocytes play an important role in extracellular K+ homeostasis in white matter and that selective loss of this channel from oligodendrocytes is sufficient to impair K+ clearance and promote seizures.
Collapse
|
46
|
Wang Y, Chen Z. An update for epilepsy research and antiepileptic drug development: Toward precise circuit therapy. Pharmacol Ther 2019; 201:77-93. [PMID: 31128154 DOI: 10.1016/j.pharmthera.2019.05.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Epilepsy involves neuronal dysfunction at molecular, cellular, and circuit levels. The understanding of the mechanism of the epilepsies has advanced greatly in the last three decades, especially in terms of their cellular and molecular basis. However, despite the availability of ~30 anti-epileptic drugs (AEDs) with diverse molecular targets, there are still many challenges (e.g. drug resistance, side effects) in pharmacological treatment of epilepsies today. Because molecular mechanisms are integrated at the level of neuronal circuits, we suggest a shift in epilepsy treatment and research strategies from the "molecular" level to the "circuit" level. Recent technological advances have facilitated circuit mechanistic discovery at each level and have paved the way for many opportunities of novel therapeutic strategies and AED development toward precise circuit therapy.
Collapse
Affiliation(s)
- Yi Wang
- Institute of Pharmacology and Toxicology, Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
47
|
Zhang F, Liu Y, Tang F, Liang B, Chen H, Zhang H, Wang K. Electrophysiological and pharmacological characterization of a novel and potent neuronal Kv7 channel opener SCR2682 for antiepilepsy. FASEB J 2019; 33:9154-9166. [DOI: 10.1096/fj.201802848rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Fan Zhang
- The Key Laboratory of Neural and Vascular Biology Ministry of Education The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province Department of Pharmacology Hebei Medical University Shijiazhuang China
| | - Yani Liu
- Department of Pharmacology Qingdao University Qingdao China
| | - Feng Tang
- Medicinal Chemistry, Simcere Pharmaceutical Nanjing China
| | - Bo Liang
- Medicinal Chemistry Shanghai Zhimeng BioPharma Shanghai China
| | - Huanming Chen
- Medicinal Chemistry Shanghai Zhimeng BioPharma Shanghai China
| | - Hailin Zhang
- The Key Laboratory of Neural and Vascular Biology Ministry of Education The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province Department of Pharmacology Hebei Medical University Shijiazhuang China
| | - Kewei Wang
- Department of Pharmacology Qingdao University Qingdao China
- Institute of Innovative Drugs School of Pharmacy Qingdao University Qingdao China
- Center for Brain Science and Brain‐Inspired Intelligence Guangzhou China
| |
Collapse
|
48
|
In Silico Analysis of the Subtype Selective Blockage of KCNA Ion Channels through the µ-Conotoxins PIIIA, SIIIA, and GIIIA. Mar Drugs 2019; 17:md17030180. [PMID: 30893914 PMCID: PMC6471588 DOI: 10.3390/md17030180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
Understanding subtype specific ion channel pore blockage by natural peptide-based toxins is crucial for developing such compounds into promising drug candidates. Herein, docking and molecular dynamics simulations were employed in order to understand the dynamics and binding states of the µ-conotoxins, PIIIA, SIIIA, and GIIIA, at the voltage-gated potassium channels of the KV1 family, and they were correlated with their experimental activities recently reported by Leipold et al. Their different activities can only adequately be understood when dynamic information about the toxin-channel systems is available. For all of the channel-bound toxins investigated herein, a certain conformational flexibility was observed during the molecular dynamic simulations, which corresponds to their bioactivity. Our data suggest a similar binding mode of µ-PIIIA at KV1.6 and KV1.1, in which a plethora of hydrogen bonds are formed by the Arg and Lys residues within the α-helical core region of µ-PIIIA, with the central pore residues of the channel. Furthermore, the contribution of the K+ channel’s outer and inner pore loops with respect to the toxin binding. and how the subtype specificity is induced, were proposed.
Collapse
|
49
|
Sun H, Lin AH, Ru F, Patil MJ, Meeker S, Lee LY, Undem BJ. KCNQ/M-channels regulate mouse vagal bronchopulmonary C-fiber excitability and cough sensitivity. JCI Insight 2019; 4:124467. [PMID: 30721152 DOI: 10.1172/jci.insight.124467] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/29/2019] [Indexed: 01/06/2023] Open
Abstract
Increased airway vagal sensory C-fiber activity contributes to the symptoms of inflammatory airway diseases. The KCNQ/Kv7/M-channel is a well-known determinant of neuronal excitability, yet whether it regulates the activity of vagal bronchopulmonary C-fibers and airway reflex sensitivity remains unknown. Here we addressed this issue using single-cell RT-PCR, patch clamp technique, extracellular recording of single vagal nerve fibers innervating the mouse lungs, and telemetric recording of cough in free-moving mice. Single-cell mRNA analysis and biophysical properties of M-current (IM) suggest that KCNQ3/Kv7.3 is the major M-channel subunit in mouse nodose neurons. The M-channel opener retigabine negatively shifted the voltage-dependent activation of IM, leading to membrane hyperpolarization, increased rheobase, and suppression of both evoked and spontaneous action potential (AP) firing in nodose neurons in an M-channel inhibitor XE991-sensitive manner. Retigabine also markedly suppressed the α,β-methylene ATP-induced AP firing in nodose C-fiber terminals innervating the mouse lungs, and coughing evoked by irritant gases in awake mice. In conclusion, KCNQ/M-channels play a role in regulating the excitability of vagal airway C-fibers at both the cell soma and nerve terminals. Drugs that open M-channels in airway sensory afferents may relieve the sufferings associated with pulmonary inflammatory diseases such as chronic coughing.
Collapse
Affiliation(s)
- Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - An-Hsuan Lin
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Fei Ru
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mayur J Patil
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lu-Yuan Lee
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
50
|
Dupont C, Denman KS, Hawash AA, Voss AA, Rich MM. Treatment of myotonia congenita with retigabine in mice. Exp Neurol 2019; 315:52-59. [PMID: 30738808 DOI: 10.1016/j.expneurol.2019.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/10/2019] [Accepted: 02/06/2019] [Indexed: 01/08/2023]
Abstract
Patients with myotonia congenita suffer from muscle stiffness caused by muscle hyperexcitability. Although loss-of-function mutations in the ClC-1 muscle chloride channel have been known for 25 years to cause myotonia congenita, this discovery has led to little progress on development of therapy. Currently, treatment is primarily focused on reducing hyperexcitability by blocking Na+ current. However, other approaches such as increasing K+ currents might also be effective. For example, the K+ channel activator retigabine, which opens KCNQ channels, is effective in treating epilepsy because it causes hyperpolarization of the resting membrane potential in neurons. In this study, we found that retigabine greatly reduced the duration of myotonia in vitro. Detailed study of its mechanism of action revealed that retigabine had no effect on any of the traditional measures of muscle excitability such as resting potential, input resistance or the properties of single action potentials. Instead it appears to shorten myotonia by activating K+ current during trains of action potentials. Retigabine also greatly reduced the severity of myotonia in vivo, which was measured using a muscle force transducer. Despite its efficacy in vivo, retigabine did not improve motor performance of mice with myotonia congenita. There are a number of potential explanations for the lack of motor improvement in vivo including central nervous system side effects. Nonetheless, the striking effectiveness of retigabine on muscle itself suggests that activating potassium currents is an effective method to treat disorders of muscle hyperexcitability.
Collapse
Affiliation(s)
- Chris Dupont
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, United States
| | - Kirsten S Denman
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, United States
| | - Ahmed A Hawash
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, United States
| | - Andrew A Voss
- Department of Biological Sciences, Wright State University, Dayton, OH 45435, United States
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, United States.
| |
Collapse
|