1
|
Lee IC, Yang YY, Chang HK, Wong SH, Yang SB. Biophysical and structural mechanisms of epilepsy-associated mutations in the S4-S5 Linker of KCNQ2 channels. Channels (Austin) 2025; 19:2464735. [PMID: 39971736 PMCID: PMC11845087 DOI: 10.1080/19336950.2025.2464735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025] Open
Abstract
Mutations in KCNQ2 are linked to various neurological disorders, including neonatal-onset epilepsy. The severity of these conditions often correlates with the mutation's location and the biochemical properties of the altered amino acid side chains. Two mutations affecting aspartate at position 212 (D212) in the S4-S5 linker of KCNQ2 have been identified. Interestingly, while the charge-conserved D212E mutation leads to severe neonatal-onset developmental and epileptic encephalopathy (DEE), the more dramatic substitution to glycine (D212G) results in self-limited familial neonatal epilepsy (SLFNE), a much milder pathology. To elucidate the underlying mechanisms, we performed electrophysiological studies and in silico simulations to investigate these mutations' biophysical and structural effects. Our findings reveal that the D212E mutation stabilizes the channel in the voltage sensor down-state and destabilizes the up-state, leading to a rightward shift in the voltage-dependent activation curve, slower activation kinetics, and accelerated deactivation kinetics. This disruption in KCNQ2 voltage sensitivity persists even in the more physiologically relevant KCNQ2/3 heterotetrameric channels. In contrast, the D212G mutation primarily destabilizes the up-state, but its impact on voltage sensitivity is significantly reduced in KCNQ2/3 heterotetrameric channels. These findings provide key insights into the biophysical and structural basis of KCNQ2 D212 mutations and their contribution to epilepsy-related symptoms, offering a clearer understanding of how these mutations drive the varied clinical outcomes observed in patients.
Collapse
Affiliation(s)
- Inn-Chi Lee
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Pediatric Neurology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | | | - Hsueh-Kai Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Swee-Hee Wong
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Pediatric Neurology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
2
|
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
|
3
|
Losgott T, Kudlacek O, Yang JW, Schicker KW, Boehm S, Salzer I. The paracetamol metabolite N-acetyl-4-benzoquinoneimine (NAPQI) prevents modulation of K V7 channels via G-protein coupled receptors by interference with PIP 2 and Ca 2+ sensitivity. Br J Pharmacol 2025; 182:1341-1357. [PMID: 39627952 DOI: 10.1111/bph.17419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Paracetamol has been found to alleviate inflammatory pain by modulating KV7 channels. Its metabolite N-acetyl-4-benzoquinoneimine (NAPQI) increases currents through these channels via a stretch of three cysteine residues in the channel S2-S3 linker. Through this effect, the excitability of neurons in the pain pathway is dampened. Inflammatory mediators, in turn, enhance the excitability of sensory neurons by inhibiting KV7 channels. Here, a specific interaction between NAPQI and the so-called inflammatory soup was investigated. EXPERIMENTAL APPROACH Currents through KV7 channels were measured in sensory neurons and after heterologous expression in tsA201 cells. In addition, changes in cytosolic Ca2+ and in the distribution of PIP2 (PI(4,5)P2) between membrane and cytosol were determined by fluorescence microscopy. KEY RESULTS NAPQI abolished Ca2+-mediated inhibitory effects of an 'inflammatory soup' containing ADP, ATP, bradykinin, histamine, 5-hydroxytryptamine, prostaglandin E2, substance P and a PAR2 agonist on KV7 channel currents in sensory neurons. Moreover, the increase of KV7.2 channel currents by quenching of cytosolic Ca2+ as well as the current decrease by depletion of membrane PIP2 was impaired by NAPQI. These effects were lost in mutant channels lacking the three cysteines in the S2-S3 linker. CONCLUSION AND IMPLICATION NAPQI targets the three-cysteine motif in the S2-S3 linker of KV7.2 channels to counteract the signalling cascades employed by inflammatory mediators that inhibit these channels. In sensory neurons, this abolishes the closure of KV7 channels by the inflammatory soup. This mechanism is likely involved in the alleviation of inflammatory pain by paracetamol.
Collapse
Affiliation(s)
- Thomas Losgott
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus W Schicker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Nappi M, Alberini G, Berselli A, Roscioni A, Soldovieri MV, Servettini I, Barrese V, Weckhuysen S, Chiu TGA, Scheffer IE, Benfenati F, Maragliano L, Miceli F, Taglialatela M. Constitutive opening of the Kv7.2 pore activation gate causes KCNQ2-developmental encephalopathy. Proc Natl Acad Sci U S A 2024; 121:e2412388121. [PMID: 39602259 PMCID: PMC11626135 DOI: 10.1073/pnas.2412388121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Pathogenic variants in KCNQ2 encoding Kv7.2 voltage-gated potassium channel subunits cause developmental encephalopathies (KCNQ2-encephalopathies), both with and without epilepsy. We herein describe the clinical, in vitro, and in silico features of two encephalopathy-causing variants (A317T, L318V) in Kv7.2 affecting two consecutive residues in the S6 activation gate that undergoes large structural rearrangements during pore opening; the disease-causing A356T variant in KCNQ3, paralogous to the A317T variant in KCNQ2, was also investigated. Currents through KCNQ2 mutant channels displayed increased density, hyperpolarizing shifts in activation gating, faster activation and slower deactivation kinetics, and resistance to changes in the cellular concentrations of phosphatidylinositol 4,5-bisphosphate (PIP2), a critical regulator of Kv7 channel function; all these features are consistent with a strong gain-of-function effect. An increase in the probability of single-channel opening, with no change in membrane abundance or single-channel conductance, was responsible for the observed gain-of-function effects. All-atom molecular dynamics simulations revealed that the mutations widened the inner pore gate and stabilized a constitutively open channel configuration in the closed state, with minimal effects on the open conformation. Thus, mutation-induced stabilization of the inner pore gate open configuration is a molecular pathogenetic mechanism for KCNQ2-related encephalopathies.
Collapse
Affiliation(s)
- Mario Nappi
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Giulio Alberini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova16132, Italy
| | - Alessandro Berselli
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Experimental Medicine, Università degli Studi di Genova, Genova16132, Italy
| | - Agnese Roscioni
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona60131, Italy
| | | | - Ilenio Servettini
- Department of Medicine and Health Science, University of Molise, Campobasso86100, Italy
| | - Vincenzo Barrese
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, Antwerp2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp2610, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp2610, Belgium
| | - Ting-Gee Annie Chiu
- Division of Medicine, Dentistry and Health Sciences, University of Melbourne, Austin Health, Melbourne, VIC3084, Australia
| | - Ingrid E. Scheffer
- The Florey Institute of Neuroscience and Mental Health and Murdoch Children’s Research Institutes, University of Melbourne, Austin and Royal Children’s Hospital, Melbourne, VIC3052, Australia
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova16132, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona60131, Italy
| | - Francesco Miceli
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Maurizio Taglialatela
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| |
Collapse
|
5
|
Ma L, Ma R, Ran R, Li J, Pan X, Guo Z, Lin X, Wen D, Wu S, Chen Y. Novel associations between KCNQ1 rs231840 polymorphism and preeclampsia in Chinese gestational women: A case-control candidate genetic study. Medicine (Baltimore) 2024; 103:e39778. [PMID: 39465874 PMCID: PMC11479463 DOI: 10.1097/md.0000000000039778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024] Open
Abstract
Preeclampsia is a complex disorder with genetic and environmental interactions. In this study, we analyzed the associations of KCNQ1gene polymorphisms with preeclampsia in Chinese pregnant women. The 3 candidate single-nucleotide polymorphisms rs231840, rs2237892, and rs2237895 were genotyped in this case-control study; clinical and biochemical data were included and SNPs were gathered from 248 individuals with preeclampsia and 237 controls. The TT genotype rs231840 increased the risk of preeclampsia (OR: 1.633; 95% CI: 1.027-2.597) and was associated with higher blood glucose levels. The haplotype TCA containing the allele of rs231840 (T), rs2237892 (C), and rs2237895 (A) was highly protective against preeclampsia and associated with the levels of blood glucose in preeclamptic patients. A novel function was found for the haplotype CCA in SNPs rs231840 (C), rs2237892 (C), and rs2237895 (A); it might be a protective combination against preeclampsia. The KCNQ1 (TT) genotype seems to be associated with preeclampsia and might affect the regulation of blood glucose in Chinese pregnant women.
Collapse
Affiliation(s)
- Lingyu Ma
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| | - Rui Ma
- Department of Obstetrics, Maternal and Child Health Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Ran Ran
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| | - Jiaying Li
- Department of Research and Development, Yinfeng Biological Engineering Technology Company Limited, Jilin, China
| | - Xuefeng Pan
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| | - Zhiheng Guo
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| | - Xichen Lin
- Affiliated Middle School to Jilin University, Jilin, China
| | - Dezhong Wen
- Department of Medical Genetics, College of Basic Medical Sciences, Jilin University, China
| | - Shuyao Wu
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| | - Ying Chen
- Department of Gynecology and Obstetrics Center, the First Hospital of Jilin University, Jilin, China
| |
Collapse
|
6
|
Wong SH, Liou YM, Yang JJ, Lee IC. KCNQ2 mutations cause unique neonatal behavior arrests without motor seizures: Functional characterization. Epilepsy Behav 2024; 156:109798. [PMID: 38788659 DOI: 10.1016/j.yebeh.2024.109798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/15/2024] [Accepted: 04/14/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE KCNQ2 gene mutation usually manifests as neonatal seizures in the first week of life. Nonsense mutations cause a unique self-limited familial neonatal epilepsy (SLFNE), which is radically different from developmental epileptic encephalopathy (DEE). However, the exact underlying mechanisms remain unclear. METHODS The proband, along with their mother and grandmother, carried the c.1342C > T (p.Arg448Ter) mutation in the KCNQ2 gene. The clinical phenotypes, electroencephalography (EEG) findings, and neurodevelopmental outcomes were comprehensively surveyed. The mutant variants were transfected into HEK293 cells to investigate functional changes. RESULTS The proband exhibited behavior arrests, autonomic and non-motor neonatal seizures with changes in heart rate and respiration. EEG exhibited focal sharp waves. Seizures were remitted after three months of age. The neurodevelopmental outcomes at three years of age were unremarkable. A functional study demonstrated that the currents of p.Arg448Ter were non-functional in homomeric p.Arg448Ter compared with that of the KCNQ2 wild type. However, the current density and V1/2 exhibited significant improvement and close to that of the wild-type after transfection with heteromeric KCNQ2 + p.Arg448Ter and KCNQ2 + KCNQ3 + p.Arg448Ter respectively. Channel expression on the cell membrane was not visible after homomeric transfection, but not after heteromeric transfection. Retigabine did not affect homomeric p.Arg448Ter but improved heteromeric p. Arg448Ter + KCNQ2 and heteromeric KCNQ2 + Arg448Ter + KCNQ3. CONCLUSIONS The newborn carrying the p. Arg448Ter mutation presented frequent behavioral arrests, autonomic, and non-motor neonatal seizures. This unique pattern differs from KCNQ2 seizures, which typically manifest as motor seizures. Although p.Arg448Ter is a non-sense decay, the functional study demonstrated an almost-full compensation mechanism after transfection of heteromeric KCNQ2 and KCNQ3.
Collapse
Affiliation(s)
- Swee-Hee Wong
- Division of Pediatric Neurology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Center, Rong Hsing Research Center for Translational Medicine, Natinal Chung Hsing University, Taichung 40227, Taiwan
| | - Jiann-Jou Yang
- Genetics Laboratory and Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Inn-Chi Lee
- Division of Pediatric Neurology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
7
|
Krüger J, Lerche H. Retigabine and gabapentin restore channel function and neuronal firing in a cellular model of an epilepsy-associated dominant-negative KCNQ5 variant. Neuropharmacology 2024; 250:109892. [PMID: 38428481 DOI: 10.1016/j.neuropharm.2024.109892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/19/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
KCNQ5 encodes the voltage-gated potassium channel KV7.5, a member of the KV7 channel family, which conducts the M-current. This current is a potent regulator of neuronal excitability by regulating membrane potential in the subthreshold range of action potentials and mediating the medium and slow afterhyperpolarization. Recently, we have identified five loss-of-function variants in KCNQ5 in patients with genetic generalized epilepsy. Using the most severe dominant-negative variant (R359C), we set out to investigate pharmacological therapeutic intervention by KV7 channel openers on channel function and neuronal firing. Retigabine and gabapentin increased R359C-derived M-current amplitudes in HEK cells expressing homomeric or heteromeric mutant KV7.5 channels. Retigabine was most effective in restoring K+ currents. Ten μM retigabine was sufficient to reach the level of WT currents without retigabine, whereas 100 μM of gabapentin showed less than half of this effect and application of 50 μM ZnCl2 only significantly increased M-current amplitude in heteromeric channels. Overexpression of KV7.5-WT potently inhibited neuronal firing by increasing the M-current, whereas R359C overexpression had the opposite effect and additionally decreased the medium afterhyperpolarization current. Both aforementioned drugs and Zn2+ reversed the effect of R359C expression by reducing firing to nearly normal levels at high current injections. Our study shows that a dominant-negative variant with a complete loss-of-function in KV7.5 leads to largely increased neuronal firing which may explain a neuronal hyperexcitability in patients. KV7 channel openers, such as retigabine or gabapentin, could be treatment options for patients currently displaying pharmacoresistant epilepsy and carrying loss-of-function variants in KCNQ5.
Collapse
Affiliation(s)
- Johanna Krüger
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Straße 27, 72076, Tübingen, Germany.
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Straße 27, 72076, Tübingen, Germany.
| |
Collapse
|
8
|
Stott JB, Greenwood IA. G protein βγ regulation of KCNQ-encoded voltage-dependent K channels. Front Physiol 2024; 15:1382904. [PMID: 38655029 PMCID: PMC11035767 DOI: 10.3389/fphys.2024.1382904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The KCNQ family is comprised of five genes and the expression products form voltage-gated potassium channels (Kv7.1-7.5) that have a major impact upon cellular physiology in many cell types. Each functional Kv7 channel forms as a tetramer that often associates with proteins encoded by the KCNE gene family (KCNE1-5) and is critically reliant upon binding of phosphatidylinositol bisphosphate (PIP2) and calmodulin. Other modulators like A-kinase anchoring proteins, ubiquitin ligases and Ca-calmodulin kinase II alter Kv7 channel function and trafficking in an isoform specific manner. It has now been identified that for Kv7.4, G protein βγ subunits (Gβγ) can be added to the list of key regulators and is paramount for channel activity. This article provides an overview of this nascent field of research, highlighting themes and directions for future study.
Collapse
Affiliation(s)
| | - Iain A. Greenwood
- Vascular Biology Research Group, Institute of Molecular and Clinical Sciences, St George’s University of London, London, United Kingdom
| |
Collapse
|
9
|
Delgado-Ramírez M, López-Serrano AL, Sánchez-Armass S, Meza U, Rodríguez-Menchaca AA. Crosstalk between cholesterol and PIP 2 in the regulation of Kv7.2/Kv7.3 channels. Biol Chem 2024; 405:161-165. [PMID: 37552610 DOI: 10.1515/hsz-2023-0204] [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: 05/05/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023]
Abstract
The activity of neuronal Kv7.2/Kv7.3 channels is critically dependent on PIP2 and finely modulated by cholesterol. Here, we report the crosstalk between cholesterol and PIP2 in the regulation of Kv7.2/Kv7.3 channels. Our results show that currents passing through Kv7.2/Kv7.3 channels in cholesterol-depleted cells, by acute application of methyl-β-cyclodextrin (MβCD), were less sensitive to PIP2 dephosphorylation strategies than those of control cells, suggesting that cholesterol depletion enhances the Kv7.2/Kv7.3-PIP2 interaction. In contrast, the sensitivity of Kv7.2/Kv7.3 channels to acute membrane cholesterol depletion by MβCD was not altered in mutant channels with different apparent affinities for PIP2.
Collapse
Affiliation(s)
- Mayra Delgado-Ramírez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, México
| | - Ana Laura López-Serrano
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, México
| | - Sergio Sánchez-Armass
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, México
| | - Ulises Meza
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, México
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, México
| |
Collapse
|
10
|
Fedida D, Sastre D, Dou Y, Westhoff M, Eldstrom J. Evaluating sequential and allosteric activation models in IKs channels with mutated voltage sensors. J Gen Physiol 2024; 156:e202313465. [PMID: 38294435 PMCID: PMC10829594 DOI: 10.1085/jgp.202313465] [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: 08/16/2023] [Revised: 11/30/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
The ion-conducting IKs channel complex, important in cardiac repolarization and arrhythmias, comprises tetramers of KCNQ1 α-subunits along with 1-4 KCNE1 accessory subunits and calmodulin regulatory molecules. The E160R mutation in individual KCNQ1 subunits was used to prevent activation of voltage sensors and allow direct determination of transition rate data from complexes opening with a fixed number of 1, 2, or 4 activatable voltage sensors. Markov models were used to test the suitability of sequential versus allosteric models of IKs activation by comparing simulations with experimental steady-state and transient activation kinetics, voltage-sensor fluorescence from channels with two or four activatable domains, and limiting slope currents at negative potentials. Sequential Hodgkin-Huxley-type models approximately describe IKs currents but cannot explain an activation delay in channels with only one activatable subunit or the hyperpolarizing shift in the conductance-voltage relationship with more activatable voltage sensors. Incorporating two voltage sensor activation steps in sequential models and a concerted step in opening via rates derived from fluorescence measurements improves models but does not resolve fundamental differences with experimental data. Limiting slope current data that show the opening of channels at negative potentials and very low open probability are better simulated using allosteric models of activation with one transition per voltage sensor, which implies that movement of all four sensors is not required for IKs conductance. Tiered allosteric models with two activating transitions per voltage sensor can fully account for IKs current and fluorescence activation kinetics in constructs with different numbers of activatable voltage sensors.
Collapse
Affiliation(s)
- David Fedida
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Daniel Sastre
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Ying Dou
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Maartje Westhoff
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Jodene Eldstrom
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
11
|
del Rivero Morfin PJ, Kochiss AL, Liedl KR, Flucher BE, Fernández-Quintero ML, Ben-Johny M. Asymmetric contribution of a selectivity filter gate in triggering inactivation of CaV1.3 channels. J Gen Physiol 2024; 156:e202313365. [PMID: 38175169 PMCID: PMC10771039 DOI: 10.1085/jgp.202313365] [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: 02/01/2023] [Revised: 10/08/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Voltage-dependent and Ca2+-dependent inactivation (VDI and CDI, respectively) of CaV channels are two biologically consequential feedback mechanisms that fine-tune Ca2+ entry into neurons and cardiomyocytes. Although known to be initiated by distinct molecular events, how these processes obstruct conduction through the channel pore remains poorly defined. Here, focusing on ultrahighly conserved tryptophan residues in the interdomain interfaces near the selectivity filter of CaV1.3, we demonstrate a critical role for asymmetric conformational changes in mediating VDI and CDI. Specifically, mutagenesis of the domain III-IV interface, but not others, enhanced VDI. Molecular dynamics simulations demonstrate that mutations in distinct selectivity filter interfaces differentially impact conformational flexibility. Furthermore, mutations in distinct domains preferentially disrupt CDI mediated by the N- versus C-lobes of CaM, thus uncovering a scheme of structural bifurcation of CaM signaling. These findings highlight the fundamental importance of the asymmetric arrangement of the pseudotetrameric CaV pore domain for feedback inhibition.
Collapse
Affiliation(s)
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
12
|
Rodrigues TCML, Dias AL, dos Santos AMF, Messias Monteiro AF, Oliveira MCN, Oliveira Pires HF, de Sousa NF, Salvadori MGDSS, Scotti MT, Scotti L. Multi-target Phenylpropanoids Against Epilepsy. Curr Neuropharmacol 2024; 22:2168-2190. [PMID: 38847378 PMCID: PMC11337686 DOI: 10.2174/1570159x22666240524160126] [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: 07/31/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 06/13/2024] Open
Abstract
Epilepsy is a neurological disease with no defined cause, characterized by recurrent epileptic seizures. These occur due to the dysregulation of excitatory and inhibitory neurotransmitters in the central nervous system (CNS). Psychopharmaceuticals have undesirable side effects; many patients require more than one pharmacotherapy to control crises. With this in mind, this work emphasizes the discovery of new substances from natural products that can combat epileptic seizures. Using in silico techniques, this review aims to evaluate the antiepileptic and multi-target activity of phenylpropanoid derivatives. Initially, ligand-based virtual screening models (LBVS) were performed with 468 phenylpropanoid compounds to predict biological activities. The LBVS were developed for the targets alpha- amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), voltage-gated calcium channel Ttype (CaV), gamma-aminobutyric acid A (GABAA), gamma-aminobutyric acid transporter type 1 (GAT-1), voltage-gated potassium channel of the Q family (KCNQ), voltage-gated sodium channel (NaV), and N-methyl D-aspartate (NMDA). The compounds that had good results in the LBVS were analyzed for the absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters, and later, the best molecules were evaluated in the molecular docking consensus. The TR430 compound showed the best results in pharmacokinetic parameters; its oral absorption was 99.03%, it did not violate any Lipinski rule, it showed good bioavailability, and no cytotoxicity was observed either from the molecule or from the metabolites in the evaluated parameters. TR430 was able to bind with GABAA (activation) and AMPA (inhibition) targets and demonstrated good binding energy and significant interactions with both targets. The studied compound showed to be a promising molecule with a possible multi-target activity in both fundamental pharmacological targets for the treatment of epilepsy.
Collapse
Affiliation(s)
| | - Arthur Lins Dias
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Aline Matilde Ferreira dos Santos
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Alex France Messias Monteiro
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | - Mayara Cecile Nascimento Oliveira
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Hugo Fernandes Oliveira Pires
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Natália Ferreira de Sousa
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | | | - Marcus Tullius Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
- Teaching and Research Management, University Hospital Lauro Wanderley, Federal University of Paraíba, 58050-585, João Pessoa, PB, Brazil
| |
Collapse
|
13
|
Archer C. Isothermal Titration Calorimetry for Fragment-Based Analysis of Ion Channel Interactions. Methods Mol Biol 2024; 2796:271-289. [PMID: 38856907 DOI: 10.1007/978-1-0716-3818-7_16] [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] [Indexed: 06/11/2024]
Abstract
Ion channels are membrane proteins that may also have intracellular and extracellular domains that interact with other ligands. In many cases, these interaction sites are highly mobile and may undergo changes in the configuration upon binding with regulatory signaling molecules. Isothermal titration calorimetry (ITC) is a powerful technique to quantify protein-ligand interactions of purified samples in solution. This chapter describes a fragment-based analysis method using ITC to quantify the interactions between a domain of the voltage-gated Kv7 channel and the calcium-regulated protein calmodulin. This example can be used to quantify the interactions between specific domains of other ion channels and their regulatory signaling proteins.
Collapse
Affiliation(s)
- Crystal Archer
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
14
|
Shorthouse D, Zhuang L, Rahrmann EP, Kosmidou C, Wickham Rahrmann K, Hall M, Greenwood BM, Devonshire G, Gilbertson RJ, Fitzgerald RC, Hall BA. KCNQ potassium channels modulate Wnt activity in gastro-oesophageal adenocarcinomas. Life Sci Alliance 2023; 6:e202302124. [PMID: 37748809 PMCID: PMC10520261 DOI: 10.26508/lsa.202302124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Voltage-sensitive potassium channels play an important role in controlling membrane potential and ionic homeostasis in the gut and have been implicated in gastrointestinal (GI) cancers. Through large-scale analysis of 897 patients with gastro-oesophageal adenocarcinomas (GOAs) coupled with in vitro models, we find KCNQ family genes are mutated in ∼30% of patients, and play therapeutically targetable roles in GOA cancer growth. KCNQ1 and KCNQ3 mediate the WNT pathway and MYC to increase proliferation through resultant effects on cadherin junctions. This also highlights novel roles of KCNQ3 in non-excitable tissues. We also discover that activity of KCNQ3 sensitises cancer cells to existing potassium channel inhibitors and that inhibition of KCNQ activity reduces proliferation of GOA cancer cells. These findings reveal a novel and exploitable role of potassium channels in the advancement of human cancer, and highlight that supplemental treatments for GOAs may exist through KCNQ inhibitors.
Collapse
Affiliation(s)
- David Shorthouse
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| | - Lizhe Zhuang
- Institute for Early Detection, CRUK Cambridge Centre, Cambridge, UK
| | - Eric P Rahrmann
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | | | - Michael Hall
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Benedict M Greenwood
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| | - Ginny Devonshire
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Richard J Gilbertson
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| |
Collapse
|
15
|
del Rivero Morfin PJ, Kochiss AL, Liedl KR, Flucher BE, Fernández-Quintero ML, Ben-Johny M. Asymmetric Contribution of a Selectivity Filter Gate in Triggering Inactivation of Ca V1.3 Channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558864. [PMID: 37790368 PMCID: PMC10542529 DOI: 10.1101/2023.09.21.558864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Voltage-dependent and Ca2+-dependent inactivation (VDI and CDI, respectively) of CaV channels are two biologically consequential feedback mechanisms that fine-tune Ca2+ entry into neurons and cardiomyocytes. Although known to be initiated by distinct molecular events, how these processes obstruct conduction through the channel pore remains poorly defined. Here, focusing on ultra-highly conserved tryptophan residues in the inter-domain interfaces near the selectivity filter of CaV1.3, we demonstrate a critical role for asymmetric conformational changes in mediating VDI and CDI. Specifically, mutagenesis of the domain III-IV interface, but not others, enhanced VDI. Molecular dynamics simulations demonstrate that mutations in distinct selectivity filter interfaces differentially impact conformational flexibility. Furthermore, mutations in distinct domains preferentially disrupt CDI mediated by the N- versus C-lobes of CaM, thus uncovering a scheme of structural bifurcation of CaM signaling. These findings highlight the fundamental importance of the asymmetric arrangement of the pseudo-tetrameric CaV pore domain for feedback inhibition.
Collapse
Affiliation(s)
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | | | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University
| |
Collapse
|
16
|
Manville RW, Hogenkamp D, Abbott GW. Ancient medicinal plant rosemary contains a highly efficacious and isoform-selective KCNQ potassium channel opener. Commun Biol 2023; 6:644. [PMID: 37322081 PMCID: PMC10272180 DOI: 10.1038/s42003-023-05021-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
Voltage-gated potassium (Kv) channels in the KCNQ subfamily serve essential roles in the nervous system, heart, muscle and epithelia. Different heteromeric KCNQ complexes likely serve distinct functions in the brain but heteromer subtype-specific small molecules for research or therapy are lacking. Rosemary (Salvia rosmarinus) is an evergreen plant used medicinally for millennia for neurological and other disorders. Here, we report that rosemary extract is a highly efficacious opener of heteromeric KCNQ3/5 channels, with weak effects on KCNQ2/3. Using functional screening we find that carnosic acid, a phenolic diterpene from rosemary, is a potent, highly efficacious, PIP2 depletion-resistant KCNQ3 opener with lesser effects on KCNQ5 and none on KCNQ1 or KCNQ2. Carnosic acid is also highly selective for KCNQ3/5 over KCNQ2/3 heteromers. Medicinal chemistry, in silico docking, and mutagenesis reveal that carboxylate-guanidinium ionic bonding with an S4-5 linker arginine underlies the KCNQ3 opening proficiency of carnosic acid, the effects of which on KCNQ3/5 suggest unique therapeutic potential and a molecular basis for ancient neurotherapeutic use of rosemary.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Derk Hogenkamp
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
17
|
Vasileva V, Chubinskiy-Nadezhdin V. Regulation of PIEZO1 channels by lipids and the structural components of extracellular matrix/cell cytoskeleton. J Cell Physiol 2023; 238:918-930. [PMID: 36947588 DOI: 10.1002/jcp.31001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
PIEZO1 is a mechanosensitive channel widely presented in eukaryotic organisms. Although the PIEZO family was discovered in 2010, main questions related to the molecular structure as well as to specific activation mechanisms and regulating pathways remain open. Two hypotheses of PIEZO1 gating were formulated: the first, as a dominant hypothesis, through the plasma membrane (force-from-lipids) and the second, via the participation of the cytoskeleton and the components of the extracellular matrix (ECM) (force-from-filaments). Many researchers provide convincing evidence for both hypotheses. It was demonstrated that PIEZO1 has a propeller-like shape forming a membrane curvature within the lipid bilayer. That suggests the participation of lipids in channel modulation, and many studies demonstrate the critical role of lipids and compounds that modify the lipid bilayer in the regulation of PIEZO1 properties. At the same time, the components of ECM and cortical cytoskeleton can be affected by the membrane curvature and thus have an impact on PIEZO1 properties. In living cells, PIEZO1 properties are reported to be critically dependent on channel microenvironment that is on combinatorial influence of plasma membrane, cytoskeleton and ECM. Thus, it is necessary to understand which factors can affect PIEZO1 and consider them when interpreting the role of PIEZO1 in various physiological processes. This review summarizes the current knowledge about regulation of Piezo1 by lipids and the components of ECM and cytoskeleton.
Collapse
Affiliation(s)
- Valeria Vasileva
- Group of Ionic Mechanisms of Cell Signalling, Department of Intracellular Signalling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Vladislav Chubinskiy-Nadezhdin
- Group of Ionic Mechanisms of Cell Signalling, Department of Intracellular Signalling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
18
|
Chokvithaya S, Caengprasath N, Buasong A, Jantasuwan S, Santawong K, Leela-adisorn N, Tongkobpetch S, Ittiwut C, Saengow VE, Kamolvisit W, Boonsimma P, Bongsebandhu-phubhakdi S, Shotelersuk V. Nine patients with KCNQ2-related neonatal seizures and functional studies of two missense variants. Sci Rep 2023; 13:3328. [PMID: 36849527 PMCID: PMC9971330 DOI: 10.1038/s41598-023-29924-y] [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: 08/16/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Mutations in KCNQ2 encoding for voltage-gated K channel subunits underlying the neuronal M-current have been associated with infantile-onset epileptic disorders. The clinical spectrum ranges from self-limited neonatal seizures to epileptic encephalopathy and delayed development. Mutations in KCNQ2 could be either gain- or loss-of-function which require different therapeutic approaches. To better understand genotype-phenotype correlation, more reports of patients and their mutations with elucidated molecular mechanism are needed. We studied 104 patients with infantile-onset pharmacoresistant epilepsy who underwent exome or genome sequencing. Nine patients with neonatal-onset seizures from unrelated families were found to harbor pathogenic or likely pathogenic variants in the KCNQ2 gene. The p.(N258K) was recently reported, and p. (G279D) has never been previously reported. Functional effect of p.(N258K) and p.(G279D) has never been previously studied. The cellular localization study demonstrated that the surface membrane expression of Kv7.2 carrying either variant was decreased. Whole-cell patch-clamp analyses revealed that both variants significantly impaired Kv7.2 M-current amplitude and density, conductance depolarizing shift in voltage dependence of activation, membrane resistance, and membrane time constant (Tau), indicating a loss-of-function in both the homotetrameric and heterotetrameric with Kv7.3 channels. In addition, both variants exerted dominant-negative effects in heterotetrameric with Kv7.3 channels. This study expands the mutational spectrum of KCNQ2- related epilepsy and their functional consequences provide insights into their pathomechanism.
Collapse
Affiliation(s)
- Suphalak Chokvithaya
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand ,grid.415584.90000 0004 0576 1386Department of Clinical Pathology and Medical Technology Laboratory, Queen Sirikit National Institute of Child Health, Ministry of Public Health, Bangkok, Thailand
| | - Natarin Caengprasath
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Aayalida Buasong
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Supavadee Jantasuwan
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Kanokwan Santawong
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Netchanok Leela-adisorn
- grid.7922.e0000 0001 0244 7875Department of Stem Cell and Cell, Therapy Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Siraprapa Tongkobpetch
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Chupong Ittiwut
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Vitchayaporn Emarach Saengow
- grid.416297.f0000 0004 0388 8201Department of Pediatrics, Maharat Nakhon Ratchasima Hospital, Nakhon Ratchasima, Thailand
| | - Wuttichart Kamolvisit
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| | - Ponghatai Boonsimma
- Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand. .,Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand.
| | - Saknan Bongsebandhu-phubhakdi
- grid.7922.e0000 0001 0244 7875Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Chula Neuroscience Center, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- grid.7922.e0000 0001 0244 7875Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.419934.20000 0001 1018 2627Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330 Thailand
| |
Collapse
|
19
|
Kongmeneck AD, Kasimova MA, Tarek M. Modulation of the IKS channel by PIP2 requires two binding sites per monomer. BBA ADVANCES 2023; 3:100073. [PMID: 37082259 PMCID: PMC10074941 DOI: 10.1016/j.bbadva.2023.100073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The phosphatidyl-inositol-4,5-bisphosphate (PIP2) lipid has been shown to be crucial for the coupling between the voltage sensor and the pore of the potassium voltage-gated KV7 channel family, especially the KV7.1 channel. Expressed in the myocardium membrane, KV7.1 forms a complex with KCNE1 auxiliary subunits to generate the IKS current. Here we present molecular models of the transmembrane region of this complex in its three known states, namely the Resting/Closed (RC), the Intermediate/Closed (IC), and the Activated/Open (AO), robustness of which is assessed by agreement with a range of biophysical data. Molecular Dynamics (MD) simulations of these models embedded in a lipid bilayer including phosphatidyl-inositol-4,5-bisphosphate (PIP2) lipids show that in presence of KCNE1, two PIP2 lipids are necessary to stabilize each state. The simulations also show that KCNE1 interacts with both PIP2 binding sites, forming a tourniquet around the pore and preventing its opening. The present investigation provides therefore key molecular elements that govern the role of PIP2 in KCNE1 modulation of IKS channels, possibly a common mechanism by which auxiliary KCNE subunits might modulate a variety of other ion channels.
Collapse
|
20
|
Yang X, Chen S, Zhang S, Shi S, Zong R, Gao Y, Guan B, Gamper N, Gao H. Intracellular zinc protects Kv7 K + channels from Ca 2+/calmodulin-mediated inhibition. J Biol Chem 2022; 299:102819. [PMID: 36549648 PMCID: PMC9852549 DOI: 10.1016/j.jbc.2022.102819] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Zinc (Zn) is an essential trace element; it serves as a cofactor for a great number of enzymes, transcription factors, receptors, and other proteins. Zinc is also an important signaling molecule, which can be released from intracellular stores into the cytosol or extracellular space, for example, during synaptic transmission. Amongst cellular effects of zinc is activation of Kv7 (KCNQ, M-type) voltage-gated potassium channels. Here, we investigated relationships between Kv7 channel inhibition by Ca2+/calmodulin (CaM) and zinc-mediated potentiation. We show that Zn2+ ionophore, zinc pyrithione (ZnPy), can prevent or reverse Ca2+/CaM-mediated inhibition of Kv7.2. In the presence of both Ca2+ and Zn2+, the Kv7.2 channels lose most of their voltage dependence and lock in an open state. In addition, we demonstrate that mutations that interfere with CaM binding to Kv7.2 and Kv7.3 reduced channel membrane abundance and activity, but these mutants retained zinc sensitivity. Moreover, the relative efficacy of ZnPy to activate these mutants was generally greater, compared with the WT channels. Finally, we show that zinc sensitivity was retained in Kv7.2 channels assembled with mutant CaM with all four EF hands disabled, suggesting that it is unlikely to be mediated by CaM. Taken together, our findings indicate that zinc is a potent Kv7 stabilizer, which may protect these channels from physiological inhibitory effects of neurotransmitters and neuromodulators, protecting neurons from overactivity.
Collapse
Affiliation(s)
- Xinhe Yang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China,CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co, Ltd, Shijiazhuang, Hebei, China
| | - Shuai Chen
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuo Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sai Shi
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, China
| | - Rui Zong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiting Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bingcai Guan
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China; Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.
| | - Haixia Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
21
|
Emerging mechanisms involving brain Kv7 channel in the pathogenesis of hypertension. Biochem Pharmacol 2022; 206:115318. [PMID: 36283445 DOI: 10.1016/j.bcp.2022.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Hypertension is a prevalent health problem inducing many organ damages. The pathogenesis of hypertension involves a complex integration of different organ systems including the brain. The elevated sympathetic nerve activity is closely related to the etiology of hypertension. Ion channels are critical regulators of neuronal excitability. Several mechanisms have been proposed to contribute to hypothalamic-driven elevated sympathetic activity, including altered ion channel function. Recent findings indicate one of the voltage-gated potassium channels, Kv7 channels (M channels), plays a vital role in regulating cardiovascular-related neurons activity, and the expression of Kv7 channels is downregulated in hypertension. This review highlights recent findings that the Kv7 channels in the brain, blood vessels, and kidneys are emerging targets involved in the pathogenesis of hypertension, suggesting new therapeutic targets for treating drug-resistant, neurogenic hypertension.
Collapse
|
22
|
Miceli F, Millevert C, Soldovieri MV, Mosca I, Ambrosino P, Carotenuto L, Schrader D, Lee HK, Riviello J, Hong W, Risen S, Emrick L, Amin H, Ville D, Edery P, de Bellescize J, Michaud V, Van-Gils J, Goizet C, Willemsen MH, Kleefstra T, Møller RS, Bayat A, Devinsky O, Sands T, Korenke GC, Kluger G, Mefford HC, Brilstra E, Lesca G, Milh M, Cooper EC, Taglialatela M, Weckhuysen S. KCNQ2 R144 variants cause neurodevelopmental disability with language impairment and autistic features without neonatal seizures through a gain-of-function mechanism. EBioMedicine 2022; 81:104130. [PMID: 35780567 PMCID: PMC9254340 DOI: 10.1016/j.ebiom.2022.104130] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 01/10/2023] Open
Abstract
Background Prior studies have revealed remarkable phenotypic heterogeneity in KCNQ2-related disorders, correlated with effects on biophysical features of heterologously expressed channels. Here, we assessed phenotypes and functional properties associated with KCNQ2 missense variants R144W, R144Q, and R144G. We also explored in vitro blockade of channels carrying R144Q mutant subunits by amitriptyline. Methods Patients were identified using the RIKEE database and through clinical collaborators. Phenotypes were collected by a standardized questionnaire. Functional and pharmacological properties of variant subunits were analyzed by whole-cell patch-clamp recordings. Findings Detailed clinical information on fifteen patients (14 novel and 1 previously published) was analyzed. All patients had developmental delay with prominent language impairment. R144Q patients were more severely affected than R144W patients. Infantile to childhood onset epilepsy occurred in 40%, while 67% of sleep-EEGs showed sleep-activated epileptiform activity. Ten patients (67%) showed autistic features. Activation gating of homomeric Kv7.2 R144W/Q/G channels was left-shifted, suggesting gain-of-function effects. Amitriptyline blocked channels containing Kv7.2 and Kv7.2 R144Q subunits. Interpretation Patients carrying KCNQ2 R144 gain-of-function variants have developmental delay with prominent language impairment, autistic features, often accompanied by infantile- to childhood-onset epilepsy and EEG sleep-activated epileptiform activity. The absence of neonatal seizures is a robust and important clinical differentiator between KCNQ2 gain-of-function and loss-of-function variants. The Kv7.2/7.3 channel blocker amitriptyline might represent a targeted treatment. Funding Supported by FWO, GSKE, KCNQ2-Cure, Jack Pribaz Foundation, European Joint Programme on Rare Disease 2020, the Italian Ministry for University and Research, the Italian Ministry of Health, the European Commission, the University of Antwerp, NINDS, and Chalk Family Foundation.
Collapse
|
23
|
KCNQ2 Selectivity Filter Mutations Cause Kv7.2 M-Current Dysfunction and Configuration Changes Manifesting as Epileptic Encephalopathies and Autistic Spectrum Disorders. Cells 2022; 11:cells11050894. [PMID: 35269516 PMCID: PMC8909571 DOI: 10.3390/cells11050894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 02/01/2023] Open
Abstract
KCNQ2 mutations can cause benign familial neonatal convulsions (BFNCs), epileptic encephalopathy (EE), and mild-to-profound neurodevelopmental disabilities. Mutations in the KCNQ2 selectivity filter (SF) are critical to neurodevelopmental outcomes. Three patients with neonatal EE carry de novo heterozygous KCNQ2 p.Thr287Ile, p.Gly281Glu and p.Pro285Thr, and all are followed-up in our clinics. Whole-cell patch-clamp analysis with transfected mutations was performed. The Kv7.2 in three mutations demonstrated significant current changes in the homomeric-transfected cells. The conduction curves for V1/2, the K slope, and currents in 3 mutations were lower than those for the wild type (WT). The p.Gly281Glu had a worse conductance than the p.Thr287Ile and p.Pro285Thr, the patient compatible with p.Gly281Glu had a worse clinical outcome than patients with p.Thr287Ile and p.Pro285Thr. The p.Gly281Glu had more amino acid weight changes than the p.Gly281Glu and p.Pro285Thr. Among 5 BFNCs and 23 EE from mutations in the SF, the greater weight of the mutated protein compared with that of the WT was presumed to cause an obstacle to pore size, which is one of the most important factors in the phenotype and outcome. For the 35 mutations in the SF domain, using changes in amino acid weight between the WT and the KCNQ2 mutations to predict EE resulted in 80.0% sensitivity and 80% specificity, a positive prediction rate of 96.0%, and a negative prediction rate of 40.0% (p = 0.006, χ2 (1, n = 35) = 7.56; odds ratio 16.0, 95% confidence interval, 1.50 to 170.63). The findings suggest that p.Thr287Ile, p.Gly281Glu and p.Pro285Thr are pathogenic to KCNQ2 EE. In mutations in SF, a mutated protein heavier than the WT is a factor in the Kv7.2 current and outcome.
Collapse
|
24
|
Maltan L, Andova AM, Derler I. The Role of Lipids in CRAC Channel Function. Biomolecules 2022; 12:biom12030352. [PMID: 35327543 PMCID: PMC8944985 DOI: 10.3390/biom12030352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/12/2022] [Accepted: 02/20/2022] [Indexed: 11/28/2022] Open
Abstract
The composition and dynamics of the lipid membrane define the physical properties of the bilayer and consequently affect the function of the incorporated membrane transporters, which also applies for the prominent Ca2+ release-activated Ca2+ ion channel (CRAC). This channel is activated by receptor-induced Ca2+ store depletion of the endoplasmic reticulum (ER) and consists of two transmembrane proteins, STIM1 and Orai1. STIM1 is anchored in the ER membrane and senses changes in the ER luminal Ca2+ concentration. Orai1 is the Ca2+-selective, pore-forming CRAC channel component located in the plasma membrane (PM). Ca2+ store-depletion of the ER triggers activation of STIM1 proteins, which subsequently leads to a conformational change and oligomerization of STIM1 and its coupling to as well as activation of Orai1 channels at the ER-PM contact sites. Although STIM1 and Orai1 are sufficient for CRAC channel activation, their efficient activation and deactivation is fine-tuned by a variety of lipids and lipid- and/or ER-PM junction-dependent accessory proteins. The underlying mechanisms for lipid-mediated CRAC channel modulation as well as the still open questions, are presented in this review.
Collapse
|
25
|
Bivalent recognition of fatty acyl-CoA by a human integral membrane palmitoyltransferase. Proc Natl Acad Sci U S A 2022; 119:2022050119. [PMID: 35140179 PMCID: PMC8851515 DOI: 10.1073/pnas.2022050119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 11/18/2022] Open
Abstract
Protein palmitoylation is one of the most highly abundant protein modifications, through which long-chain fatty acids get attached to cysteines by a thioester linkage. It plays critically important roles in growth signaling, the organization of synaptic receptors, and the regulation of ion channel function. Yet the molecular mechanism of the DHHC family of integral membrane enzymes that catalyze this modification remains poorly understood. Here, we present the structure of a precatalytic complex of human DHHC20 with palmitoyl CoA. Together with the accompanying functional data, the structure shows how a bivalent recognition of palmitoyl CoA by the DHHC enzyme, simultaneously at both the fatty acyl group and the CoA headgroup, is essential for catalytic chemistry to proceed. S-acylation, also known as palmitoylation, is the most abundant form of protein lipidation in humans. This reversible posttranslational modification, which targets thousands of proteins, is catalyzed by 23 members of the DHHC family of integral membrane enzymes. DHHC enzymes use fatty acyl-CoA as the ubiquitous fatty acyl donor and become autoacylated at a catalytic cysteine; this intermediate subsequently transfers the fatty acyl group to a cysteine in the target protein. Protein S-acylation intersects with almost all areas of human physiology, and several DHHC enzymes are considered as possible therapeutic targets against diseases such as cancer. These efforts would greatly benefit from a detailed understanding of the molecular basis for this crucial enzymatic reaction. Here, we combine X-ray crystallography with all-atom molecular dynamics simulations to elucidate the structure of the precatalytic complex of human DHHC20 in complex with palmitoyl CoA. The resulting structure reveals that the fatty acyl chain inserts into a hydrophobic pocket within the transmembrane spanning region of the protein, whereas the CoA headgroup is recognized by the cytosolic domain through polar and ionic interactions. Biochemical experiments corroborate the predictions from our structural model. We show, using both computational and experimental analyses, that palmitoyl CoA acts as a bivalent ligand where the interaction of the DHHC enzyme with both the fatty acyl chain and the CoA headgroup is important for catalytic chemistry to proceed. This bivalency explains how, in the presence of high concentrations of free CoA under physiological conditions, DHHC enzymes can efficiently use palmitoyl CoA as a substrate for autoacylation.
Collapse
|
26
|
Sanguinetti MC, Seebohm G. Physiological Functions, Biophysical Properties, and Regulation of KCNQ1 (K V7.1) Potassium Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:335-353. [PMID: 35138621 DOI: 10.1007/978-981-16-4254-8_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
KCNQ1 (KV7.1) K+ channels are expressed in multiple tissues, including the heart, pancreas, colon, and inner ear. The gene encoding the KCNQ1 protein was discovered by a positional cloning effort to determine the genetic basis of long QT syndrome, an inherited ventricular arrhythmia that can cause sudden death. Mutations in KCNQ1 can also cause other types of arrhythmia (i.e., short QT syndrome, atrial fibrillation) and the gene may also have a role in diabetes and certain cancers. KCNQ1 α-subunits can partner with accessory β-subunits (KCNE1-KCNE5) to form K+-selective channels that have divergent biophysical properties. In the heart, KCNQ1 α-subunits coassemble with KCNE1 β-subunits to form channels that conduct IKs, a very slowly activating delayed rectifier K+ current. KV7.1 channels are highly regulated by PIP2, calmodulin, and phosphorylation, and rich pharmacology includes blockers and gating modulators. Recent biophysical studies and a cryo-EM structure of the KCNQ1-calmodulin complex have provided new insights into KV7.1 channel function, and how interactions between KCNQ1 and KCNE subunits alter the gating properties of heteromultimeric channels.
Collapse
Affiliation(s)
| | - Guiscard Seebohm
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases, University Hospital Münster, Münster, Germany
| |
Collapse
|
27
|
Kawai T, Okamura Y. Spotlight on the Binding Affinity of Ion Channels for Phosphoinositides: From the Study of Sperm Flagellum. Front Physiol 2022; 13:834180. [PMID: 35197868 PMCID: PMC8859416 DOI: 10.3389/fphys.2022.834180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/07/2022] [Indexed: 11/21/2022] Open
Abstract
The previous studies revealed that many types of ion channels have sensitivity to PtdIns(4,5)P2, which has been mainly shown using heterologous expression system. On the other hand, there remains few evidence showing that PtdIns(4,5)P2 natively regulate the ion channel activities in physiological context. Our group recently discovered that a sperm specific K+ channel, Slo3, is natively regulated by PtdIns(4,5)P2 in sperm flagellum. Very interestingly, a principal piece, to which Slo3 specifically localized, had extremely low density of PtdIns(4,5)P2 compared to the regular cell plasma membrane. Furthermore, our studies and the previous ones also revealed that Slo3 had much stronger PtdIns(4,5)P2 affinity than KCNQ2/3 channels, which are widely regulated by endogenous PtdIns(4,5)P2 in neurons. Thus, the high-PtdIns(4,5)P2 affinity of Slo3 is well-adapted to the specialized PtdIns(4,5)P2 environment in the principal piece. This study sheds light on the relationship between PtdIns(4,5)P2-affinity of ion channels and their PtdIns(4,5)P2 environment in native cells. We discuss the current understanding about PtdIns(4,5)P2 affinity of diverse ion channels and their possible regulatory mechanism in native cellular environment.
Collapse
Affiliation(s)
- Takafumi Kawai
- Integrative Physiology Program, Graduate School of Medicine, Osaka University, Suita, Japan
- *Correspondence: Takafumi Kawai,
| | - Yasushi Okamura
- Integrative Physiology Program, Graduate School of Medicine, Osaka University, Suita, Japan
- Graduate School of Frontier Bioscience, Osaka University, Suita, Japan
| |
Collapse
|
28
|
Riel EB, Jürs BC, Cordeiro S, Musinszki M, Schewe M, Baukrowitz T. The versatile regulation of K2P channels by polyanionic lipids of the phosphoinositide and fatty acid metabolism. J Gen Physiol 2022; 154:212926. [PMID: 34928298 PMCID: PMC8693234 DOI: 10.1085/jgp.202112989] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/01/2021] [Indexed: 12/29/2022] Open
Abstract
Work over the past three decades has greatly advanced our understanding of the regulation of Kir K+ channels by polyanionic lipids of the phosphoinositide (e.g., PIP2) and fatty acid metabolism (e.g., oleoyl-CoA). However, comparatively little is known regarding the regulation of the K2P channel family by phosphoinositides and by long-chain fatty acid–CoA esters, such as oleoyl-CoA. We screened 12 mammalian K2P channels and report effects of polyanionic lipids on all tested channels. We observed activation of members of the TREK, TALK, and THIK subfamilies, with the strongest activation by PIP2 for TRAAK and the strongest activation by oleoyl-CoA for TALK-2. By contrast, we observed inhibition for members of the TASK and TRESK subfamilies. Our results reveal that TASK-2 channels have both activatory and inhibitory PIP2 sites with different affinities. Finally, we provided evidence that PIP2 inhibition of TASK-1 and TASK-3 channels is mediated by closure of the recently identified lower X-gate as critical mutations within the gate (i.e., L244A, R245A) prevent PIP2-induced inhibition. Our findings establish that K+ channels of the K2P family are highly sensitive to polyanionic lipids, extending our knowledge of the mechanisms of lipid regulation and implicating the metabolism of these lipids as possible effector pathways to regulate K2P channel activity.
Collapse
Affiliation(s)
- Elena B Riel
- Institute of Physiology, Kiel University, Kiel, Germany
| | - Björn C Jürs
- Institute of Physiology, Kiel University, Kiel, Germany.,Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | | | | | - Marcus Schewe
- Institute of Physiology, Kiel University, Kiel, Germany
| | | |
Collapse
|
29
|
Zheng Y, Liu H, Chen Y, Dong S, Wang F, Wang S, Li GL, Shu Y, Xu F. Structural insights into the lipid and ligand regulation of a human neuronal KCNQ channel. Neuron 2021; 110:237-247.e4. [PMID: 34767770 DOI: 10.1016/j.neuron.2021.10.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 01/10/2023]
Abstract
The KCNQ family (KCNQ1-KCNQ5) of voltage-gated potassium channels plays critical roles in many physiological and pathological processes. It is known that the channel opening of all KCNQs relies on the signaling lipid molecule phosphatidylinositol 4,5-bisphosphate (PIP2). However, the molecular mechanism of PIP2 in modulating the opening of the four neuronal KCNQ channels (KCNQ2-KCNQ5), which are essential for regulating neuronal excitability, remains largely elusive. Here, we report the cryoelectron microscopy (cryo-EM) structures of human KCNQ4 determined in complex with the activator ML213 in the absence or presence of PIP2. Two PIP2 molecules are identified in the open-state structure of KCNQ4, which act as a bridge to couple the voltage-sensing domain (VSD) and pore domain (PD) of KCNQ4 leading to the channel opening. Our findings reveal the binding sites and activation mechanisms of ML213 and PIP2 for neuronal KCNQ channels, providing a framework for therapeutic intervention targeting on these important channels.
Collapse
Affiliation(s)
- You Zheng
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Heng Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuxin Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Shaowei Dong
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Fang Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Shengyi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Geng-Lin Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China.
| | - Fei Xu
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
PIP 2-dependent coupling of voltage sensor and pore domains in K v7.2 channel. Commun Biol 2021; 4:1189. [PMID: 34650221 PMCID: PMC8517023 DOI: 10.1038/s42003-021-02729-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/23/2021] [Indexed: 01/10/2023] Open
Abstract
Phosphatidylinositol-4,5-bisphosphate (PIP2) is a signaling lipid which regulates voltage-gated Kv7/KCNQ potassium channels. Altered PIP2 sensitivity of neuronal Kv7.2 channel is involved in KCNQ2 epileptic encephalopathy. However, the molecular action of PIP2 on Kv7.2 gating remains largely elusive. Here, we use molecular dynamics simulations and electrophysiology to characterize PIP2 binding sites in a human Kv7.2 channel. In the closed state, PIP2 localizes to the periphery of the voltage-sensing domain (VSD). In the open state, PIP2 binds to 4 distinct interfaces formed by the cytoplasmic ends of the VSD, the gate, intracellular helices A and B and their linkers. PIP2 binding induces bilayer-interacting conformation of helices A and B and the correlated motion of the VSD and the pore domain, whereas charge-neutralizing mutations block this coupling and reduce PIP2 sensitivity of Kv7.2 channels by disrupting PIP2 binding. These findings reveal the allosteric role of PIP2 in Kv7.2 channel activation. Pant et al. describe the mechanism by which PIP2 might regulate homomeric Kv7.2 channels. They identify sites important in the binding of the PIP2 lipid to Kv7.2 channels and propose that the PIP2 binding to a specific site results in the coupling between the voltage sensor domain (VSD) and pore domain (PD), which stabilizes the open state of the channel.
Collapse
|
31
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
32
|
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
|
33
|
Pipatpolkai T, Quetschlich D, Stansfeld PJ. From Bench to Biomolecular Simulation: Phospholipid Modulation of Potassium Channels. J Mol Biol 2021; 433:167105. [PMID: 34139216 PMCID: PMC8361781 DOI: 10.1016/j.jmb.2021.167105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/05/2022]
Abstract
Potassium (K+) ion channels are crucial in numerous cellular processes as they hyperpolarise a cell through K+ conductance, returning a cell to its resting potential. K+ channel mutations result in multiple clinical complications such as arrhythmia, neonatal diabetes and migraines. Since 1995, the regulation of K+ channels by phospholipids has been heavily studied using a range of interdisciplinary methods such as cellular electrophysiology, structural biology and computational modelling. As a result, K+ channels are model proteins for the analysis of protein-lipid interactions. In this review, we will focus on the roles of lipids in the regulation of K+ channels, and how atomic-level structures, along with experimental techniques and molecular simulations, have helped guide our understanding of the importance of phospholipid interactions.
Collapse
Affiliation(s)
- Tanadet Pipatpolkai
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK; OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PT, UK
| | - Daniel Quetschlich
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Chemistry, South Parks Road, Oxford OX1 3QZ, UK
| | - Phillip J Stansfeld
- School of Life Sciences & Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
34
|
Abstract
Neurons are highly specialized cells equipped with a sophisticated molecular machinery for the reception, integration, conduction and distribution of information. The evolutionary origin of neurons remains unsolved. How did novel and pre-existing proteins assemble into the complex machinery of the synapse and of the apparatus conducting current along the neuron? In this review, the step-wise assembly of functional modules in neuron evolution serves as a paradigm for the emergence and modification of molecular machinery in the evolution of cell types in multicellular organisms. The pre-synaptic machinery emerged through modification of calcium-regulated large vesicle release, while the postsynaptic machinery has different origins: the glutamatergic postsynapse originated through the fusion of a sensory signaling module and a module for filopodial outgrowth, while the GABAergic postsynapse incorporated an ancient actin regulatory module. The synaptic junction, in turn, is built around two adhesion modules controlled by phosphorylation, which resemble septate and adherens junctions. Finally, neuronal action potentials emerged via a series of duplications and modifications of voltage-gated ion channels. Based on these origins, key molecular innovations are identified that led to the birth of the first neuron in animal evolution.
Collapse
|
35
|
Novel KCNQ4 variants in different functional domains confer genotype- and mechanism-based therapeutics in patients with nonsyndromic hearing loss. Exp Mol Med 2021; 53:1192-1204. [PMID: 34316018 PMCID: PMC8333092 DOI: 10.1038/s12276-021-00653-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Loss-of-function variant in the gene encoding the KCNQ4 potassium channel causes autosomal dominant nonsyndromic hearing loss (DFNA2), and no effective pharmacotherapeutics have been developed to reverse channel activity impairment. Phosphatidylinositol 4,5-bisphosphate (PIP2), an obligatory phospholipid for maintaining KCNQ channel activity, confers differential pharmacological sensitivity of channels to KCNQ openers. Through whole-exome sequencing of DFNA2 families, we identified three novel KCNQ4 variants related to diverse auditory phenotypes in the proximal C-terminus (p.Arg331Gln), the C-terminus of the S6 segment (p.Gly319Asp), and the pore region (p.Ala271_Asp272del). Potassium currents in HEK293T cells expressing each KCNQ4 variant were recorded by patch-clamp, and functional recovery by PIP2 expression or KCNQ openers was examined. In the homomeric expression setting, the three novel KCNQ4 mutant proteins lost conductance and were unresponsive to KCNQ openers or PIP2 expression. Loss of p.Arg331Gln conductance was slightly restored by a tandem concatemer channel (WT-p.R331Q), and increased PIP2 expression further increased the concatemer current to the level of the WT channel. Strikingly, an impaired homomeric p.Gly319Asp channel exhibited hyperactivity when a concatemer (WT-p.G319D), with a negative shift in the voltage dependence of activation. Correspondingly, a KCNQ inhibitor and chelation of PIP2 effectively downregulated the hyperactive WT-p.G319D concatemer channel. Conversely, the pore-region variant (p.Ala271_Asp272del) was nonrescuable under any condition. Collectively, these novel KCNQ4 variants may constitute therapeutic targets that can be manipulated by the PIP2 level and KCNQ-regulating drugs under the physiological context of heterozygous expression. Our research contributes to the establishment of a genotype/mechanism-based therapeutic portfolio for DFNA2.
Collapse
|
36
|
Abstract
K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).
Collapse
|
37
|
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
|
38
|
Wilson ZT, Jiang M, Geng J, Kaur S, Workman SW, Hao J, Bernas T, Tseng GN. Delayed KCNQ1/KCNE1 assembly on the cell surface helps I Ks fulfil its function as a repolarization reserve in the heart. J Physiol 2021; 599:3337-3361. [PMID: 33963564 DOI: 10.1113/jp281773] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In adult ventricular myocytes, the slow delayed rectifier (IKs ) channels are distributed on the surface sarcolemma, not t-tubules. In adult ventricular myocytes, KCNQ1 and KCNE1 have distinct cell surface and cytoplasmic pools. KCNQ1 and KCNE1 traffic from the endoplasmic reticulum to the plasma membrane by separate routes, and assemble into IKs channels on the cell surface. Liquid chromatography/tandem mass spectrometry applied to affinity-purified KCNQ1 and KCNE1 interacting proteins reveals novel interactors involved in protein trafficking and assembly. Microtubule plus-end binding protein 1 (EB1) binds KCNQ1 preferentially in its dimer form, and promotes KCNQ1 to reach the cell surface. An LQT1-associated mutation, Y111C, reduces KCNQ1 binding to EB1 dimer. ABSTRACT Slow delayed rectifier (IKs ) channels consist of KCNQ1 and KCNE1. IKs functions as a 'repolarization reserve' in the heart by providing extra current for ventricular action potential shortening during β-adrenergic stimulation. There has been much debate about how KCNQ1 and KCNE1 traffic in cells, where they associate to form IKs channels, and the distribution pattern of IKs channels relative to β-adrenergic signalling complex. We used experimental strategies not previously applied to KCNQ1, KCNE1 or IKs , to provide new insights into these issues. 'Retention-using-selected-hook' experiments showed that newly translated KCNE1 constitutively trafficked through the conventional secretory path to the cell surface. KCNQ1 largely stayed in the endoplasmic reticulum, although dynamic KCNQ1 vesicles were observed in the submembrane region. Disulphide-bonded KCNQ1/KCNE1 constructs reported preferential association after they had reached cell surface. An in situ proximity ligation assay detected IKs channels in surface sarcolemma but not t-tubules of ventricular myocytes, similar to the reported location of adenylate cyclase 9/yotiao. Fluorescent protein-tagged KCNQ1 and KCNE1, in conjunction with antibodies targeting their extracellular epitopes, detected distinct cell surface and cytoplasmic pools of both proteins in myocytes. We conclude that, in cardiomyocytes, KCNQ1 and KCNE1 traffic by different routes to surface sarcolemma where they assemble into IKs channels. This mode of delayed channel assembly helps IKs fulfil its function of repolarization reserve. Proteomic experiments revealed a novel KCNQ1 interactor, microtubule plus-end binding protein 1 (EB1). EB1 dimer (active form) bound KCNQ1 and increased its surface level. An LQT1 mutation, Y111C, reduced KCNQ1 binding to EB1 dimer.
Collapse
Affiliation(s)
- Zachary T Wilson
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Min Jiang
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA, USA.,Institute of Medicinal biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Geng
- Institute of Medicinal biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sukhleen Kaur
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Samuel W Workman
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA, USA.,Present address: School of Medicine, Rutgers University, Piscataway, NJ, USA
| | - Jon Hao
- Poochon Scientific, Frederick, MD, USA
| | - Tytus Bernas
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Gea-Ny Tseng
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
39
|
Tan T, Wang W, Liu T, Zhong P, Conrow-Graham M, Tian X, Yan Z. Neural circuits and activity dynamics underlying sex-specific effects of chronic social isolation stress. Cell Rep 2021; 34:108874. [PMID: 33761364 DOI: 10.1016/j.celrep.2021.108874] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/09/2020] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
Exposure to prolonged stress in critical developmental periods induces heightened vulnerability to psychiatric disorders, which may have sex-specific consequences. Here we investigate the neuronal circuits mediating behavioral changes in mice after chronic adolescent social isolation stress. Escalated aggression is exhibited in stressed males, while social withdrawal is shown in stressed females. In vivo multichannel recordings of free-moving animals indicate that pyramidal neurons in prefrontal cortex (PFC) from stressed males exhibit the significantly decreased spike activity during aggressive attacks, while PFC pyramidal neurons from stressed females show a blunted increase of discharge rates during sociability tests. Chemogenetic and electrophysiological evidence shows that PFC hypofunctioning and BLA principal neuron hyperactivity contribute to the elevated aggression in stressed males, while PFC hypofunctioning and VTA dopamine neuron hypoactivity contribute to the diminished sociability in stressed females. These results establish a framework for understanding the circuit and physiological mechanisms underlying sex-specific divergent effects of stress.
Collapse
Affiliation(s)
- Tao Tan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Wei Wang
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Tiaotiao Liu
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Megan Conrow-Graham
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Xin Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
40
|
Effertz T, Moser T, Oliver D. Recent advances in cochlear hair cell nanophysiology: subcellular compartmentalization of electrical signaling in compact sensory cells. Fac Rev 2021; 9:24. [PMID: 33659956 PMCID: PMC7886071 DOI: 10.12703/r/9-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years, genetics, physiology, and structural biology have advanced into the molecular details of the sensory physiology of auditory hair cells. Inner hair cells (IHCs) and outer hair cells (OHCs) mediate two key functions: active amplification and non-linear compression of cochlear vibrations by OHCs and sound encoding by IHCs at their afferent synapses with the spiral ganglion neurons. OHCs and IHCs share some molecular physiology, e.g. mechanotransduction at the apical hair bundles, ribbon-type presynaptic active zones, and ionic conductances in the basolateral membrane. Unique features enabling their specific function include prestin-based electromotility of OHCs and indefatigable transmitter release at the highest known rates by ribbon-type IHC active zones. Despite their compact morphology, the molecular machineries that either generate electrical signals or are driven by these signals are essentially all segregated into local subcellular structures. This review provides a brief account on recent insights into the molecular physiology of cochlear hair cells with a specific focus on organization into membrane domains.
Collapse
Affiliation(s)
- Thomas Effertz
- InnerEarLab, Department of Otorhinolaryngology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Dominik Oliver
- Institute for Physiology and Pathophysiology, Philipps University, Deutschhausstraße 2, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodelling, GRK 2213, Philipps University, Marburg, Germany
| |
Collapse
|
41
|
Abstract
Kv7.1-Kv7.5 (KCNQ1-5) K+ channels are voltage-gated K+ channels with major roles in neurons, muscle cells and epithelia where they underlie physiologically important K+ currents, such as neuronal M current and cardiac IKs. Specific biophysical properties of Kv7 channels make them particularly well placed to control the activity of excitable cells. Indeed, these channels often work as 'excitability breaks' and are targeted by various hormones and modulators to regulate cellular activity outputs. Genetic deficiencies in all five KCNQ genes result in human excitability disorders, including epilepsy, arrhythmias, deafness and some others. Not surprisingly, this channel family attracts considerable attention as potential drug targets. Here we will review biophysical properties and tissue expression profile of Kv7 channels, discuss recent advances in the understanding of their structure as well as their role in various neurological, cardiovascular and other diseases and pathologies. We will also consider a scope for therapeutic targeting of Kv7 channels for treatment of the above health conditions.
Collapse
|
42
|
Li T, Wu K, Yue Z, Wang Y, Zhang F, Shen H. Structural Basis for the Modulation of Human KCNQ4 by Small-Molecule Drugs. Mol Cell 2020; 81:25-37.e4. [PMID: 33238160 DOI: 10.1016/j.molcel.2020.10.037] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/28/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Among the five KCNQ channels, also known as the Kv7 voltage-gated potassium (Kv) channels, KCNQ2-KCNQ5 control neuronal excitability. Dysfunctions of KCNQ2-KCNQ5 are associated with neurological disorders such as epilepsy, deafness, and neuropathic pain. Here, we report the cryoelectron microscopy (cryo-EM) structures of human KCNQ4 and its complexes with the opener retigabine or the blocker linopirdine at overall resolutions of 2.5, 3.1, and 3.3 Å, respectively. In all structures, a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule inserts its head group into a cavity within each voltage-sensing domain (VSD), revealing an unobserved binding mode for PIP2. Retigabine nestles in each fenestration, inducing local shifts. Instead of staying within the central pore, linopirdine resides in a cytosolic cavity underneath the inner gate. Electrophysiological analyses of various mutants corroborated the structural observations. Our studies reveal the molecular basis for the modulatory mechanism of neuronal KCNQ channels and provide a framework for structure-facilitated drug discovery targeting these important channels.
Collapse
Affiliation(s)
- Tian Li
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Kun Wu
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhenlei Yue
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Yifei Wang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Fan Zhang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Huaizong Shen
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
43
|
Lee JE, Park CH, Kang H, Ko J, Cho S, Woo J, Chae MR, Lee SW, Kim SJ, Kim J, So I. The agonistic action of URO-K10 on Kv7.4 and 7.5 channels is attenuated by co-expression of KCNE4 ancillary subunit. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:503-516. [PMID: 33093272 PMCID: PMC7585595 DOI: 10.4196/kjpp.2020.24.6.503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/02/2022]
Abstract
KCNQ family constitutes slowly-activating potassium channels among voltage-gated potassium channel superfamily. Recent studies suggested that KCNQ4 and 5 channels are abundantly expressed in smooth muscle cells, especially in lower urinary tract including corpus cavernosum and that both channels can exert membrane stabilizing effect in the tissues. In this article, we examined the electrophysiological characteristics of overexpressed KCNQ4, 5 channels in HEK293 cells with recently developed KCNQ-specific agonist. With submicromolar EC50, the drug not only increased the open probability of KCNQ4 channel but also increased slope conductance of the channel. The overall effect of the drug in whole-cell configuration was to increase maximal whole-cell conductance, to prolongate the activation process, and left-shift of the activation curve. The agonistic action of the drug, however, was highly attenuated by the co-expression of one of the β ancillary subunits of KCNQ family, KCNE4. Strong in vitro interactions between KCNQ4, 5 and KCNE4 were found through Foster Resonance Energy Transfer and co-immunoprecipitation. Although the expression levels of both KCNQ4 and KCNE4 are high in mesenteric arterial smooth muscle cells, we found that 1 μM of the agonist was sufficient to almost completely relax phenylephrine-induced contraction of the muscle strip. Significant expression of KCNQ4 and KCNE4 in corpus cavernosum together with high tonic contractility of the tissue grants highly promising relaxational effect of the KCNQ-specific agonist in the tissue.
Collapse
Affiliation(s)
- Jung Eun Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Christine Haewon Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hana Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Juyeon Ko
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - JooHan Woo
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Mee Ree Chae
- Department of Urology, Samsung Medical Center, Seoul 06351, Korea
| | - Sung Won Lee
- Department of Urology, Samsung Medical Center, Seoul 06351, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jinsung Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
44
|
Tran B, Ji ZG, Xu M, Tsuchida TN, Cooper EC. Two KCNQ2 Encephalopathy Variants in the Calmodulin-Binding Helix A Exhibit Dominant-Negative Effects and Altered PIP 2 Interaction. Front Physiol 2020; 11:1144. [PMID: 33041849 PMCID: PMC7518097 DOI: 10.3389/fphys.2020.571813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/18/2020] [Indexed: 11/13/2022] Open
Abstract
Heterozygous missense variants in KCNQ2, which encodes the potassium channel subunit Kv7.2, are among the most common genetic causes of severe neonatal-onset epileptic encephalopathy. Because about 20% of known severe Kv7.2 missense changes lie within the intracellular C-terminal region, improving understanding of the underlying pathogenic mechanisms is important. We analyzed the basis for the severe phenotypes of Kv7.2 A337T and A337G, variants in the C-terminal’s calmodulin (CaM)-binding Helix A. When expressed heterologously in mammalian cells, alone or in combination with wild type Kv7.2 or with wild type Kv7.2 and Kv7.3, both variants strongly suppressed channel currents. A337T channels expressed alone exhibited significantly reduced protein half-life and surface trafficking and co-immunoprecipitated less CaM. For both variants, increasing cellular phosphatidylinositol 4,5-bisphosphate (PIP2) by overexpression of PI(4)P5-kinase restored current densities. For both variants, the fraction of current suppressed by activation of M1 muscarinic receptors with 10 μM oxotremorine methiodide, which depletes PIP2, was less than for controls. During voltage-sensitive phosphatase-induced transient PIP2 depletion and resynthesize, potassium current inhibition and recovery kinetics were both markedly slowed. These results suggest that these variants may reduce currents by a mechanism not previously described: slowing of PIP2 migration between the bulk membrane and binding sites mediating channel electromechanical coupling. A novel Kv7.2/3-selective opener, SF0034, rescued current amplitudes. Our findings show that these two Helix A variants suppress channel current density strongly, consistent with their severe heterozygous phenotypes, implicate impairment of CaM and PIP2 regulation in KCNQ2 encephalopathy pathogenesis, and highlight the potential usefulness of selective Kv7 openers for this distinctive pathogenic mechanism and patient subgroup.
Collapse
Affiliation(s)
- Baouyen Tran
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Zhi-Gang Ji
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Mingxuan Xu
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Tammy N Tsuchida
- Departments of Pediatrics and Neurology, Children's National Medical Center, Washington, DC, United States
| | - Edward C Cooper
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,Department of Neurology, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
45
|
Baculis BC, Zhang J, Chung HJ. The Role of K v7 Channels in Neural Plasticity and Behavior. Front Physiol 2020; 11:568667. [PMID: 33071824 PMCID: PMC7530275 DOI: 10.3389/fphys.2020.568667] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
Activity-dependent persistent changes in neuronal intrinsic excitability and synaptic strength are widely thought to underlie learning and memory. Voltage-gated KCNQ/Kv7 potassium channels have been of great interest as the potential targets for memory disorders due to the beneficial effects of their antagonists in cognition. Importantly, de novo dominant mutations in their neuronal subunits KCNQ2/Kv7.2 and KCNQ3/Kv7.3 are associated with epilepsy and neurodevelopmental disorder characterized by developmental delay and intellectual disability. The role of Kv7 channels in neuronal excitability and epilepsy has been extensively studied. However, their functional significance in neural plasticity, learning, and memory remains largely unknown. Here, we review recent studies that support the emerging roles of Kv7 channels in intrinsic and synaptic plasticity, and their contributions to cognition and behavior.
Collapse
Affiliation(s)
- Brian C Baculis
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jiaren Zhang
- Department of Molecular Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Molecular Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
46
|
Shen L, Yang L, Zhang W. Multiple basic amino acid residues contribute to phosphatidic acid-mediated inhibition of rice potassium channel OsAKT2. PLANT SIGNALING & BEHAVIOR 2020; 15:1789818. [PMID: 32649276 PMCID: PMC8550199 DOI: 10.1080/15592324.2020.1789818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 06/11/2023]
Abstract
Anionic phospholipid phosphatidic acid (PA) behaves as an important second messenger involved in many cellular processes, such as development, cytoskeletal dynamics, vesicle trafficking, and stress response. Recently, it was reported that PA can directly bind with the rice Shaker K+ channel OsAKT2 to inhibit its channel activity. Two adjacent arginine residues (R644 and R645) in ANK domain were identified as a PA-binding site essential to the PA-mediated inhibition of OsAKT2. However, there may be still other PA-binding sites unidentified in OsAKT2. Here, using a PA biosensor (PAleon), we found that the exogenous PA treatment significantly increased the PA level at the plasma membrane of Xenopus oocytes which were used to express OsAKT2 for electrophysiological assays. As reported previously, exogenous PA markedly inhibited OsAKT2 K+ currents. Replacement of two adjacent basic residues (R190 and K191) in the S4 voltage sensor by glycine completely abolished the time-dependent K+ currents of OsAKT2, but this variant was insensitive to PA treatment. In addition, we also identified other two adjacent arginines (R755 and R756) located in the cytosolic domain as a PA-binding site, which were also essential to the PA-mediated inhibition of OsAKT2. These results provide a more comprehensive understanding of the PA-K+ channel interaction mechanism. Combining the findings here with the previous study, we propose that multiple basic residues (R190/K191, R644/R645, and R755/R756) in different domains of OsAKT2 contribute to PA-mediated regulation of OsAKT2.
Collapse
Affiliation(s)
- Like Shen
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Lele Yang
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Wenhua Zhang
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
47
|
PIP 2: A critical regulator of vascular ion channels hiding in plain sight. Proc Natl Acad Sci U S A 2020; 117:20378-20389. [PMID: 32764146 PMCID: PMC7456132 DOI: 10.1073/pnas.2006737117] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), has long been established as a major contributor to intracellular signaling, primarily by virtue of its role as a substrate for phospholipase C (PLC). Signaling by Gq-protein-coupled receptors triggers PLC-mediated hydrolysis of PIP2 into inositol 1,4,5-trisphosphate and diacylglycerol, which are well known to modulate vascular ion channel activity. Often overlooked, however, is the role PIP2 itself plays in this regulation. Although numerous reports have demonstrated that PIP2 is critical for ion channel regulation, how it impacts vascular function has received scant attention. In this review, we focus on PIP2 as a regulator of ion channels in smooth muscle cells and endothelial cells-the two major classes of vascular cells. We further address the concerted effects of such regulation on vascular function and blood flow control. We close with a consideration of current knowledge regarding disruption of PIP2 regulation of vascular ion channels in disease.
Collapse
|
48
|
Heteromeric Kv7.2 current changes caused by loss-of-function of KCNQ2 mutations are correlated with long-term neurodevelopmental outcomes. Sci Rep 2020; 10:13375. [PMID: 32770121 PMCID: PMC7415140 DOI: 10.1038/s41598-020-70212-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Pediatric epilepsy caused by KCNQ2 mutations can manifest benign familial neonatal convulsions (BFNC) to neonatal-onset epileptic encephalopathy (EE). Patients might manifest mild to profound neurodevelopmental disabilities. We analysed c.853C > A (P285T) and three mutations that cause KCNQ2 protein changes in the 247 position: c.740C > T (S247L), c.740C > A (S247X), and c.740C > G (S247W). S247L, S247W, and P285T cause neonatal-onset EE and poor neurodevelopmental outcomes; S247X cause BFNC and normal outcome. We investigated the phenotypes correlated with human embryonic kidney 293 (HEK293) cell functional current changes. More cell-current changes and a worse conductance curve were present in the homomeric transfected S247X than in S247L, S247W, and P285T. But in the heteromeric channel, S247L, S247W and P285T had more current impairments than did S247X. The protein expressions of S247X were nonfunctional. The outcomes were most severe in S247L and S247W, and severity was correlated with heteromeric current. Current changes were more significant in cells with homomeric S247X, but currents were “rescued” after heteromeric transfection of KCNQ2 and KCNQ3. This was not the case in cells with S247L, S247W. Our findings support that homomeric current changes are common in KCNQ2 neonatal-onset EE and KCNQ2 BFNC; however, heteromeric functional current changes are correlated with long-term neurodevelopmental outcomes.
Collapse
|
49
|
Abstract
Kv7 channels (Kv7.1-7.5) are voltage-gated K+ channels that can be modulated by five β-subunits (KCNE1-5). Kv7.1-KCNE1 channels produce the slow-delayed rectifying K+ current, IKs, which is important during the repolarization phase of the cardiac action potential. Kv7.2-7.5 are predominantly neuronally expressed and constitute the muscarinic M-current and control the resting membrane potential in neurons. Kv7.1 produces drastically different currents as a result of modulation by KCNE subunits. This flexibility allows the Kv7.1 channel to have many roles depending on location and assembly partners. The pharmacological sensitivity of Kv7.1 channels differs from that of Kv7.2-7.5 and is largely dependent upon the number of β-subunits present in the channel complex. As a result, the development of pharmaceuticals targeting Kv7.1 is problematic. This review discusses the roles and the mechanisms by which different signaling pathways affect Kv7.1 and KCNE channels and could potentially provide different ways of targeting the channel.
Collapse
Affiliation(s)
- Emely Thompson
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - Jodene Eldstrom
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - David Fedida
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| |
Collapse
|
50
|
Larsson JE, Frampton DJA, Liin SI. Polyunsaturated Fatty Acids as Modulators of K V7 Channels. Front Physiol 2020; 11:641. [PMID: 32595524 PMCID: PMC7300222 DOI: 10.3389/fphys.2020.00641] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/20/2020] [Indexed: 11/25/2022] Open
Abstract
Voltage-gated potassium channels of the KV7 family are expressed in many tissues. The physiological importance of KV7 channels is evident from specific forms of disorders linked to dysfunctional KV7 channels, including variants of epilepsy, cardiac arrhythmia and hearing impairment. Thus, understanding how KV7 channels are regulated in the body is of great interest. This Mini Review focuses on the effects of polyunsaturated fatty acids (PUFAs) on KV7 channel activity and possible underlying mechanisms of action. By summarizing reported effects of PUFAs on KV7 channels and native KV7-mediated currents, we conclude that the generally observed effect is a PUFA-induced increase in current amplitude. The increase in current is commonly associated with a shift in the voltage-dependence of channel opening and in some cases with increased maximum conductance. Auxiliary KCNE subunits, which associate with KV7 channels in certain tissues, may influence PUFA effects, though findings are conflicting. Both direct and indirect activating PUFA effects have been described, direct effects having been most extensively studied on KV7.1. The negative charge of the PUFA head-group has been identified as critical for electrostatic interaction with conserved positively charged amino acids in transmembrane segments 4 and 6. Additionally, the localization of double bonds in the PUFA tail tunes the apparent affinity of PUFAs to KV7.1. Indirect effects include those mediated by PUFA metabolites. Indirect inhibitory effects involve KV7 channel degradation and re-distribution from lipid rafts. Understanding how PUFAs regulate KV7 channels may provide insight into physiological regulation of KV7 channels and bring forth new therapeutic strategies.
Collapse
Affiliation(s)
- Johan E Larsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Damon J A Frampton
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sara I Liin
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|