1
|
Ignat LA, Tipa RO, Cehan AC, Bacârea VC. BK channels and alcohol tolerance: Insights from studies on Drosophila, nematodes, rodents and cell lines: A systematic review. MEDICINE INTERNATIONAL 2025; 5:33. [PMID: 40236633 PMCID: PMC11995379 DOI: 10.3892/mi.2025.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
Addictive disorders markedly affect the emotional, physical and financial wellbeing of individuals, placing a substantial burden on the healthcare system. With their widespread presence in the brain, large-conductance calcium and voltage-activated potassium (BK) channels play a crucial role in various aspects of neuronal function. They contribute to behavioral tolerance and are closely linked to neuronal activity and modulation through intracellular calcium levels. As such, BK channels serve as key models for investigating the mechanisms of the effects of alcohol. Investigating their role in alcohol tolerance provides a broader understanding of their physiological and pharmacological importance. The present systematic review examined the literature on the role of BK channels in alcohol tolerance and comprehensively explored the topic. For this purpose, two databases, Web of Science and PubMed, were searched, and studies published from 2000 until June, 2024 were included. After applying specific inclusion and exclusion criteria, 35 studies underwent analysis to present a chronological overview of BK channels and their relevance in alcohol tolerance development. The studies were categorized into four main groups, according to research conducted on: i) Fruit flies; ii) nematodes; iii) rodents; and iv) cell lines. Understanding the mechanisms through which alcohol interacts with these channels may help to elucidate the cellular and molecular mechanisms underlying alcohol tolerance. There is a growing interest in developing drugs that can precisely modulate BK channel activity to treat alcohol dependence and tolerance. However, additional studies are required to fully explain the complex mechanisms through which BK channels influence alcohol-related behaviors and to interpret these findings into clinical applications.
Collapse
Affiliation(s)
- Luciana Angela Ignat
- Doctoral School, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
| | - Raluca Oana Tipa
- ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
- Department of Psychiatry, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alina Cehan Cehan
- Plastic and Reconstructive Surgery, Emergency Clinical County Hospital of Targu Mures, 540136 Targu Mures, Romania
| | - Vladimir Constantin Bacârea
- Department of Scientific Research Methodology, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| |
Collapse
|
2
|
Park SB, Lee NY, Lee EY, Kim S, Lee N, Roh EC, Kim YG, Kim HJ, Jin MS, Park CS, Kim YC. Discovery of Diphenyl Ether Derivatives as Novel BK Ca Channel Activators: Structure-Activity Relationship, Cryo-EM Complex Structures, and In Vivo Animal Studies. J Med Chem 2025; 68:4259-4286. [PMID: 39947888 DOI: 10.1021/acs.jmedchem.4c02008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The BKCa channel, a large-conductance calcium-activated potassium channel, plays a crucial role in maintaining the homeostasis of the micturition cycle and airway-related functions. In this study, we optimized a novel BKCa channel activator, 4d, with a diphenyl ether structure identified from library screening. This led to the discovery of potent activators, 10b (EC50 = 0.12 μM, cell-based assay) and 51b, an orally bioavailable derivative. Compound 10b demonstrated potent in vivo efficacy in a spontaneous hypertensive rat (SHR) of urinary incontinence model, while compound 51b showed dose-dependent cough suppression efficacy with an ED50 of 11.8 mg/kg in a citric acid-induced cough model. Furthermore, we reported the cryo-electron microscopy (cryo-EM) structures of the BKCa channel in complex with 10b and 51b at resolutions of 2.8 and 3.4 Å. Based on structural analyses, we determined the binding sites and key interaction residues of 51b, which were validated via mutation studies.
Collapse
Affiliation(s)
- Soo Bin Park
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Na Young Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eun-Young Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Subin Kim
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Narasaem Lee
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eun Chae Roh
- College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31116, Republic of Korea
| | - Yoon Gyoon Kim
- College of Pharmacy, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungcheongnam-do 31116, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815, Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Mi Sun Jin
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Chul-Seung Park
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Yong-Chul Kim
- School of Life Science, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
3
|
Jia Z, Zhang G, Shi J, Cui J, Chen J. Intrinsic Opening of BK Channels Derives from Inherent Leakage in Hydrophobic Gating. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632877. [PMID: 39868145 PMCID: PMC11760684 DOI: 10.1101/2025.01.13.632877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The big potassium (BK) channels remain open with a small limiting probability of P o ~ 10-7 at minimal Ca2+ and negative voltages < -100 mV. The molecular origin and functional significance of such "intrinsic opening" are not understood. Here we combine atomistic simulations and electrophysiological experiments to show that the intrinsic opening of BK channels is an inherent property of the vapor barrier, generated by hydrophobic dewetting of the BK inner pore in the deactivated state. The vapor barrier only gives rise to a finite free energy barrier, of ~ 8 kcal/mol, and cannot completely shut down K+ flow even when the voltage sensor domains are fully deactivated. This results in the leaking currents that can be measured at negative voltages as the indication of intrinsic opening. The shallow limiting slope of P o at negative voltages results primarily from the electric field effects on the permeating ion through the vapor barrier. We further demonstrate that the vapor barrier can be perturbed by inner pore mutations and truncation of the cytosolic domains, leading to predicable changes in limiting slope measurements. Therefore, the intrinsic opening in BK channels, and possibly in other ion channels, opens up an opportunity to experimentally study hydrophobic gating. Our results further suggest that intrinsic opening in BK channels is the fundamental basis for the allosteric mechanism of activation by both voltage and Ca2+.
Collapse
Affiliation(s)
- Zhiguang Jia
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Guohui Zhang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, MO 63130, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, MO 63130, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, MO 63130, USA
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
4
|
Shen ZS, Gan J, Xu B, Chen YL, Zhang FF, Ji JW, Chen DH, Qiao Y, Tang QY, Zhang Z. The mechanism of Ca 2+-independent activation of BKCa channels in mouse inner hair cells and the crucial role of the BK channels in auditory perception. J Biol Chem 2025; 301:107970. [PMID: 39515584 PMCID: PMC11758846 DOI: 10.1016/j.jbc.2024.107970] [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: 04/23/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BK channels are expressed in mouse cochlear inner hair cells (IHCs) and exhibit Ca2+-independent activation at negative potentials. However, the mechanism underlying Ca2+-independent activation of the BK channels in mouse IHCs remains unknown. In this study, we found the BK channel expressed in IHCs contains both the stress-axis regulated exon 2 variant and an alternative splice of exon9 (alt9), which significantly shift the voltage dependence of the BK channels when coexpressed with LRRC52 in 0 [Ca2+]i. Furthermore, we discovered that mechanical force also induces negative shifts in the voltage dependence of IHC-expressed BK channels. Thus, we propose that the additive effects of mechanical force, special isoforms, and LRRC52 coexpression on voltage dependence shifts may account for the Ca2+-independent activation of the BK channel in IHC. Additionally, we found that the IHCs-specific deletion of the BK channels causes hearing damage in mice. Our study suggests a mechanism for Ca2+-independent activation in IHCs and highlights the crucial role of the BK channel in auditory perception.
Collapse
Affiliation(s)
- Zhong-Shan Shen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jun Gan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bing Xu
- Otorhinolaryngology Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Auditory Engineering Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ya-Lin Chen
- Otorhinolaryngology Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Auditory Engineering Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Fei-Fei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jun-Wei Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Dan-Hua Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yuehua Qiao
- Otorhinolaryngology Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Auditory Engineering Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Qiong-Yao Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, Medical University, Xuzhou, Jiangsu Province, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
5
|
Wang Q, Peng W, Yang Y, Wu Y, Han R, Ding T, Zhang X, Liu J, Yang J, Liu J. Proteome and ubiquitinome analyses of the brain cortex in K18- hACE2 mice infected with SARS-CoV-2. iScience 2024; 27:110602. [PMID: 39211577 PMCID: PMC11357812 DOI: 10.1016/j.isci.2024.110602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/03/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Clinical research indicates that SARS-CoV-2 infection is linked to several neurological consequences, and the virus is still spreading despite the availability of vaccinations and antiviral medications. To determine how hosts respond to SARS-CoV-2 infection, we employed LC-MS/MS to perform ubiquitinome and proteome analyses of the brain cortexes from K18-hACE2 mice in the presence and absence of SARS-CoV-2 infection. A total of 8,024 quantifiable proteins and 5,220 quantifiable lysine ubiquitination (Kub) sites in 2023 proteins were found. Glutamatergic synapse, calcium signaling pathway, and long-term potentiation may all play roles in the neurological consequences of SARS-CoV-2 infection. Then, we observed possible interactions between 26 SARS-CoV-2 proteins/E3 ubiquitin-protein ligases/deubiquitinases and several differentially expressed mouse proteins or Kub sites. We present the first description of the brain cortex ubiquitinome in K18-hACE2 mice, laying the groundwork for further investigation into the pathogenic processes and treatment options for neurological dysfunction following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Qiaochu Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Wanjun Peng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Yehong Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Rong Han
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Tao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xutong Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
6
|
Zhang G, Yang H, Wang Y, Liang H, Shi J, Cui J. Redox-dependent Cd 2+ inhibition of BK-type Ca 2+-activated K + channels. Biophys J 2024; 123:2076-2084. [PMID: 38400542 PMCID: PMC11309971 DOI: 10.1016/j.bpj.2024.02.015] [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: 11/21/2023] [Revised: 01/11/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024] Open
Abstract
Large-conductance Ca2+-activated K+ channels (BK channels) are formed by Slo1 subunits as a homotetramer. Besides Ca2+, other divalent cations, such as Cd2+, also activate BK channels when applied intracellularly by shifting the conductance-voltage relation to more negative voltages. However, we found that if the inside-out patch containing BK channels was treated with solution containing reducing agents such as dithiothreitol (DTT), then subsequent Cd2+ application completely inhibited BK currents. The DTT-dependent Cd2+ inhibition could be reversed by treating the patch with solutions containing H2O2, suggesting that a redox reaction regulates the Cd2+ inhibition of BK channels. Similar DTT-dependent Cd2+ inhibition was also observed in a mutant BK channel, Core-MT, in which the cytosolic domain of the channel is deleted, and in the proton-activated Slo3 channels but not observed in the voltage-gated Shaker K+ channels. A possible mechanism for the DTT-dependent Cd2+ inhibition is that DTT treatment breaks one or more disulfide bonds between cysteine pairs in the BK channel protein and the freed thiol groups coordinate with Cd2+ to form an ion bridge that blocks the channel or locks the channel at the closed state. However, surprisingly, none of the mutations of all cysteine residues in Slo1 affect the DTT-dependent Cd2+ inhibition. These results are puzzling, with an apparent contradiction: on one hand, a redox reaction seems to regulate Cd2+ inhibition of the channel, but on the other hand, no cysteine residue in the Slo1 subunit seems to be involved in such inhibition.
Collapse
Affiliation(s)
- Guohui Zhang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Huanghe Yang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri; Department of Biochemistry, Duke University Medical Center, Durham, North Carolina
| | - Yuyin Wang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Hongwu Liang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Jingyi Shi
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri.
| |
Collapse
|
7
|
Whelan SCM, Mutchler SM, Han A, Priestley C, Satlin LM, Kleyman TR, Shi S. Kcnma1 alternative splicing in mouse kidney: regulation during development and by dietary K + intake. Am J Physiol Renal Physiol 2024; 327:F49-F60. [PMID: 38779757 DOI: 10.1152/ajprenal.00100.2024] [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: 03/29/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
The pore-forming α-subunit of the large-conductance K+ (BK) channel is encoded by a single gene, KCNMA1. BK channel-mediated K+ secretion in the kidney is crucial for overall renal K+ homeostasis in both physiological and pathological conditions. BK channels achieve phenotypic diversity by various mechanisms, including substantial exon rearrangements at seven major alternative splicing sites. However, KCNMA1 alternative splicing in the kidney has not been characterized. The present study aims to identify the major splice variants of mouse Kcnma1 in whole kidney and distal nephron segments. We designed primers that specifically cross exons within each alternative splice site of mouse Kcnma1 and performed real-time quantitative RT-PCR (RT-qPCR) to quantify relative abundance of each splice variant. Our data suggest that Kcnma1 splice variants within mouse kidney are less diverse than in the brain. During postnatal kidney development, most Kcnma1 splice variants at site 5 and the COOH terminus increase in abundance over time. Within the kidney, the regulation of Kcnma1 alternative exon splicing within these two sites by dietary K+ loading is both site and sex specific. In microdissected distal tubules, the Kcnma1 alternative splicing profile, as well as its regulation by dietary K+, are distinctly different than in the whole kidney, suggesting segment and/or cell type specificity in Kcnma1 splicing events. Overall, our data provide evidence that Kcnma1 alternative splicing is regulated during postnatal development and may serve as an important adaptive mechanism to dietary K+ loading in mouse kidney.NEW & NOTEWORTHY We identified the major Kcnma1 splice variants that are specifically expressed in the whole mouse kidney or aldosterone-sensitive distal nephron segments. Our data suggest that Kcnma1 alternative splicing is developmentally regulated and subject to changes in dietary K+.
Collapse
Affiliation(s)
| | - Stephanie M Mutchler
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Agnes Han
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Catherine Priestley
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Shujie Shi
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
8
|
Peixoto-Neves D, Jaggar JH. Physiological functions and pathological involvement of ion channel trafficking in the vasculature. J Physiol 2024; 602:3275-3296. [PMID: 37818949 PMCID: PMC11006830 DOI: 10.1113/jp285007] [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: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
A variety of ion channels regulate membrane potential and calcium influx in arterial smooth muscle and endothelial cells to modify vascular functions, including contractility. The current (I) generated by a population of ion channels is equally dependent upon their number (N), open probability (Po) and single channel current (i), such that I = N.PO.i. A conventional view had been that ion channels traffic to the plasma membrane in a passive manner, resulting in a static surface population. It was also considered that channels assemble with auxiliary subunits prior to anterograde trafficking of the multimeric complex to the plasma membrane. Recent studies have demonstrated that physiological stimuli can regulate the surface abundance (N) of several different ion channels in arterial smooth muscle and endothelial cells to control arterial contractility. Physiological stimuli can also regulate the number of auxiliary subunits present in the plasma membrane to modify the biophysical properties, regulatory mechanisms and physiological functions of some ion channels. Furthermore, ion channel trafficking becomes dysfunctional in the vasculature during hypertension, which negatively impacts the regulation of contractility. The temporal kinetics of ion channel and auxiliary subunit trafficking can also vary depending on the signalling mechanisms and proteins involved. This review will summarize recent work that has uncovered the mechanisms, functions and pathological modifications of ion channel trafficking in arterial smooth muscle and endothelial cells.
Collapse
Affiliation(s)
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38139
| |
Collapse
|
9
|
Cordero Padilla K, Monefeldt GA, Guevárez Galán A, Marrero HG, Lloret-Torres ME, Velázquez-Marrero C. BK ZERO isoform HEK293 stably transfected cell lines differing 3'UTRs to assess miR-9 regulation. PLoS One 2024; 19:e0298966. [PMID: 38502673 PMCID: PMC10950231 DOI: 10.1371/journal.pone.0298966] [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: 09/26/2023] [Accepted: 01/30/2024] [Indexed: 03/21/2024] Open
Abstract
Research has identified the large conductance voltage- and calcium-activated potassium channel (BK) as a key regulator of neuronal excitability genetically associated to behavioral alcohol tolerance. Sensitivity to ethanol at the molecular level is characterized by acute potentiation of channel activity. BK isoforms show variations in alcohol sensitivity and are differentially distributed on the plasma membrane surface in response to prolonged exposure. MicroRNA (MiRNA) targeting of alcohol-sensitive isoforms coupled with active internalization of BK channels in response to ethanol are believed to be key in establishing homeostatic adaptations that produce persistent changes within the plasma membrane of neurons. In fact, microRNA 9 (miR-9) upregulated expression is a key event in persistent alcohol tolerance mediating acute EtOH desensitization of BK channels. The exact nature of these interactions remains a current topic of discussion. To further study the effects of miR-9 on the expression and distribution of BK channel isoforms we designed an experimental model by transfecting human BK channel isoforms ZERO heterologous constructs in human embryonic kidney cells 293 (HEK293) cells respectively expressing 2.1 (miR-9 responsive), 2.2 (unresponsive) and control (no sequence) 3'untranslated region (3'UTR) miRNA recognition sites. We used imaging techniques to characterize the stably transfected monoclonal cell lines, and electrophysiology to validate channel activity. Finally, we used immunocytochemistry to validate isoform responsiveness to miR-9. Our findings suggest the cell lines were successfully transfected to express either the 2.1 or 2.2 version of ZERO. Patch clamp recordings confirm that these channels retain their functionality and immunohistochemistry shows differential responses to miR-9, making these cells viable for use in future alcohol dependence studies.
Collapse
Affiliation(s)
- Katherine Cordero Padilla
- Department of Anatomy and Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Windsor University School of Medicine, St. Kitts, West Indies
| | - Gerardo Alvarado Monefeldt
- Department of Biology, University of Puerto Rico Cayey Campus, Cayey, Puerto Rico
- Samuel Merritt University, Oakland, California, United States of America
| | - Adriel Guevárez Galán
- Department of Anatomy and Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Department of Biology, University of Puerto Rico Bayamón Campus, Bayamón, Puerto Rico
| | - Hector G. Marrero
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Mario E. Lloret-Torres
- Department of Anatomy and Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Cristina Velázquez-Marrero
- Department of Anatomy and Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
10
|
Nakajo K, Kasuya G. Modulation of potassium channels by transmembrane auxiliary subunits via voltage-sensing domains. Physiol Rep 2024; 12:e15980. [PMID: 38503563 PMCID: PMC10950684 DOI: 10.14814/phy2.15980] [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: 12/29/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024] Open
Abstract
Voltage-gated K+ (KV ) and Ca2+ -activated K+ (KCa ) channels are essential proteins for membrane repolarization in excitable cells. They also play important physiological roles in non-excitable cells. Their diverse physiological functions are in part the result of their auxiliary subunits. Auxiliary subunits can alter the expression level, voltage dependence, activation/deactivation kinetics, and inactivation properties of the bound channel. KV and KCa channels are activated by membrane depolarization through the voltage-sensing domain (VSD), so modulation of KV and KCa channels through the VSD is reasonable. Recent cryo-EM structures of the KV or KCa channel complex with auxiliary subunits are shedding light on how these subunits bind to and modulate the VSD. In this review, we will discuss four examples of auxiliary subunits that bind directly to the VSD of KV or KCa channels: KCNQ1-KCNE3, Kv4-DPP6, Slo1-β4, and Slo1-γ1. Interestingly, their binding sites are all different. We also present some examples of how functionally critical binding sites can be determined by introducing mutations. These structure-guided approaches would be effective in understanding how VSD-bound auxiliary subunits modulate ion channels.
Collapse
Affiliation(s)
- Koichi Nakajo
- Division of Integrative Physiology, Department of PhysiologyJichi Medical UniversityShimotsukeJapan
| | - Go Kasuya
- Division of Integrative Physiology, Department of PhysiologyJichi Medical UniversityShimotsukeJapan
| |
Collapse
|
11
|
O'Connor EC, Kambara K, Bertrand D. Advancements in the use of xenopus oocytes for modelling neurological disease for novel drug discovery. Expert Opin Drug Discov 2024; 19:173-187. [PMID: 37850233 DOI: 10.1080/17460441.2023.2270902] [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: 05/31/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
INTRODUCTION Introduced about 50 years ago, the model of Xenopus oocytes for the expression of recombinant proteins has gained a broad spectrum of applications. The authors herein review the benefits brought from using this model system, with a focus on modeling neurological disease mechanisms and application to drug discovery. AREAS COVERED Using multiple examples spanning from ligand gated ion channels to transporters, this review presents, in the light of the latest publications, the benefits offered from using Xenopus oocytes. Studies range from the characterization of gene mutations to the discovery of novel treatments for disorders of the central nervous system (CNS). EXPERT OPINION Development of new drugs targeting CNS disorders has been marked by failures in the translation from preclinical to clinical studies. As progress in genetics and molecular biology highlights large functional differences arising from a single to a few amino acid exchanges, the need for drug screening and functional testing against human proteins is increasing. The use of Xenopus oocytes to enable precise modeling and characterization of clinically relevant genetic variants constitutes a powerful model system that can be used to inform various aspects of CNS drug discovery and development.
Collapse
Affiliation(s)
- Eoin C O'Connor
- Roche Pharma Research and Early Development, Neuroscience & Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | | | | |
Collapse
|
12
|
Liu H, Qu D, Cao Y, Li H, Wu X, Zhu Y, Tao J, Li Y, Cao C. TAT-Modified Martentoxin Displays Intravenous Antiseizure Activities. ACS Chem Neurosci 2024; 15:205-214. [PMID: 38112732 DOI: 10.1021/acschemneuro.3c00744] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Epilepsy is a chronic disease of brain dysfunction, which arises from imbalance between excitatory and inhibitory activities in neural circuits. Previously, we reported that peptide Martentoxin (MarTX), from scorpion Buthus martensii Karsch, displayed antiseizure activities by specifically inhibiting BK(α + β4) channel currents. Injection of MarTX into the hippocampal region of mice significantly alleviated convulsive seizures. However, intravenous injection of MarTX had no antiepileptic efficacy due to the blood-brain barrier (BBB). To address this, here, we designed cell-penetrating peptide TAT-modified MarTX, in which the linker containing three glycines was put between TAT and the N-terminus of MarTX (forming MTX-N-TAT) or between TAT and the C-terminus of MarTX (forming MTX-C-TAT), respectively. We prepared them in a large amount through Escherichia coli overexpression system and then probed their antiseizure activities. Our results indicated that intravenous injection of MTX-C-TAT showed significant therapeutic efficacy of antiseizure. It increased seizure latency, reduced the total seizure duration and the number of seizures at stages 3, 4, and 5, inhibited hippocampal neuronal hyperexcitability, and exhibited neuroprotective effects on hippocampal neurons. These studies implied that MTX-C-TAT displayed intravenous antiseizure activities properly through crossing BBB and would be a potential antiepileptic drug in the future.
Collapse
Affiliation(s)
- Huan Liu
- School of pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Dongxiao Qu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yunzhu Cao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
- Nanjing Fenglin Biotechnology Co., 2 Taixi Road, Pukou District, Nanjing 210031, China
| | - Haiting Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
- Center for Supramolecular Chemistry and Catalysis, Department of Chemistry, College of Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Xiaoyu Wu
- Center for Supramolecular Chemistry and Catalysis, Department of Chemistry, College of Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yudan Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie Tao
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yiming Li
- School of pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chunyang Cao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
13
|
Kallure GS, Pal K, Zhou Y, Lingle CJ, Chowdhury S. High-resolution structures illuminate key principles underlying voltage and LRRC26 regulation of Slo1 channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572542. [PMID: 38187713 PMCID: PMC10769243 DOI: 10.1101/2023.12.20.572542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Multi-modal regulation of Slo1 channels by membrane voltage, intracellular calcium, and auxiliary subunits enables its pleiotropic physiological functions. Our understanding of how voltage impacts Slo1 conformational dynamics and the mechanisms by which auxiliary subunits, particularly of the LRRC (Leucine Rich Repeat containing) family of proteins, modulate its voltage gating remain unresolved. Here, we used single particle cryo-electron microscopy to determine structures of human Slo1 mutants which functionally stabilize the closed pore (F315A) or the activated voltage-sensor (R207A). Our structures, obtained under calcium-free conditions, reveal that a key step in voltage-sensing by Slo1 involves a rotameric flip of the voltage-sensing charges (R210 and R213) moving them by ∼6 Å across a hydrophobic gasket. Next we obtained reconstructions of a complex of human Slo1 with the human LRRC26 (γ1) subunit in absence of calcium. Together with extensive biochemical tests, we show that the extracellular domains of γ1 form a ring of interlocked dominos that stabilizes the quaternary assembly of the complex and biases Slo1:γ1 assembly towards high stoichiometric complexes. The transmembrane helix of γ1 is kinked and tightly packed against the Slo1 voltage-sensor. We hypothesize that γ1 subunits exert relatively small effects on early steps in voltage-gating but structurally stabilize non-S4 helices of Slo1 voltage-sensor which energetically facilitate conformational rearrangements that occur late in voltage stimulated transitions.
Collapse
|
14
|
Bhebhe CN, Higham JP, Gupta RA, Raine T, Bulmer DC. K V7 but not dual small and intermediate K Ca channel openers inhibit the activation of colonic afferents by noxious stimuli. Am J Physiol Gastrointest Liver Physiol 2023; 325:G436-G445. [PMID: 37667839 PMCID: PMC10894664 DOI: 10.1152/ajpgi.00141.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
In numerous subtypes of central and peripheral neurons, small and intermediate conductance Ca2+-activated K+ (SK and IK, respectively) channels are important regulators of neuronal excitability. Transcripts encoding SK channel subunits, as well as the closely related IK subunit, are coexpressed in the soma of colonic afferent neurons with receptors for the algogenic mediators ATP and bradykinin, P2X3 and B2, highlighting the potential utility of these channels as drug targets for the treatment of abdominal pain in gastrointestinal diseases such as irritable bowel syndrome. Despite this, pretreatment with the dual SK/IK channel opener SKA-31 had no effect on the colonic afferent response to ATP, bradykinin, or noxious ramp distention of the colon. Inhibition of SK or IK channels with apamin or TRAM-34, respectively, yielded no change in spontaneous baseline afferent activity, indicating these channels are not tonically active. In contrast to its lack of effect in electrophysiological experiments, comparable concentrations of SKA-31 abolished ongoing peristaltic activity in the colon ex vivo. Treatment with the KV7 channel opener retigabine blunted the colonic afferent response to all applied stimuli. Our data therefore highlight the potential utility of KV7, but not SK/IK, channel openers as analgesic agents for the treatment of abdominal pain.NEW & NOTEWORTHY Despite marked coexpression of small (Kcnn1, Kcnn2) and intermediate (Kcnn4) conductance calcium-activated potassium channel transcripts with P2X3 (P2rx3) or bradykinin B2 (Bdkrb2) receptors in colonic sensory neurons, pharmacological activation of these channels had no effect on the colonic afferent response to ATP, bradykinin or luminal distension of the colon. This is in contrast to the robust inhibitory effect of the KV7 channel opener, retigabine.
Collapse
Affiliation(s)
- Charity N Bhebhe
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Rohit A Gupta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Ancatén-González C, Segura I, Alvarado-Sánchez R, Chávez AE, Latorre R. Ca 2+- and Voltage-Activated K + (BK) Channels in the Nervous System: One Gene, a Myriad of Physiological Functions. Int J Mol Sci 2023; 24:3407. [PMID: 36834817 PMCID: PMC9967218 DOI: 10.3390/ijms24043407] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
BK channels are large conductance potassium channels characterized by four pore-forming α subunits, often co-assembled with auxiliary β and γ subunits to regulate Ca2+ sensitivity, voltage dependence and gating properties. BK channels are abundantly expressed throughout the brain and in different compartments within a single neuron, including axons, synaptic terminals, dendritic arbors, and spines. Their activation produces a massive efflux of K+ ions that hyperpolarizes the cellular membrane. Together with their ability to detect changes in intracellular Ca2+ concentration, BK channels control neuronal excitability and synaptic communication through diverse mechanisms. Moreover, increasing evidence indicates that dysfunction of BK channel-mediated effects on neuronal excitability and synaptic function has been implicated in several neurological disorders, including epilepsy, fragile X syndrome, mental retardation, and autism, as well as in motor and cognitive behavior. Here, we discuss current evidence highlighting the physiological importance of this ubiquitous channel in regulating brain function and its role in the pathophysiology of different neurological disorders.
Collapse
Affiliation(s)
- Carlos Ancatén-González
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ignacio Segura
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Rosangelina Alvarado-Sánchez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Doctorado en Ciencias Mención Biofísica y Biología Computacional, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Andrés E. Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| |
Collapse
|
16
|
Potassium channelopathies associated with epilepsy-related syndromes and directions for therapeutic intervention. Biochem Pharmacol 2023; 208:115413. [PMID: 36646291 DOI: 10.1016/j.bcp.2023.115413] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
A number of mutations to members of several CNS potassium (K) channel families have been identified which result in rare forms of neonatal onset epilepsy, or syndromes of which one prominent characteristic is a form of epilepsy. Benign Familial Neonatal Convulsions or Seizures (BFNC or BFNS), also referred to as Self-Limited Familial Neonatal Epilepsy (SeLNE), results from mutations in 2 members of the KV7 family (KCNQ) of K channels; while generally self-resolving by about 15 weeks of age, these mutations significantly increase the probability of generalized seizure disorders in the adult, in some cases they result in more severe developmental syndromes. Epilepsy of Infancy with Migrating Focal Seizures (EIMSF), or Migrating Partial Seizures of Infancy (MMPSI), is a rare severe form of epilepsy linked primarily to gain of function mutations in a member of the sodium-dependent K channel family, KCNT1 or SLACK. Finally, KCNMA1 channelopathies, including Liang-Wang syndrome (LIWAS), are rare combinations of neurological symptoms including seizure, movement abnormalities, delayed development and intellectual disabilities, with Liang-Wang syndrome an extremely serious polymalformative syndrome with a number of neurological sequelae including epilepsy. These are caused by mutations in the pore-forming subunit of the large-conductance calcium-activated K channel (BK channel) KCNMA1. The identification of these rare but significant channelopathies has resulted in a resurgence of interest in their treatment by direct pharmacological or genetic modulation. We will briefly review the genetics, biophysics and pharmacology of these K channels, their linkage with the 3 syndromes described above, and efforts to more effectively target these syndromes.
Collapse
|
17
|
Sun L, Horrigan FT. A gating lever and molecular logic gate that couple voltage and calcium sensor activation to opening in BK potassium channels. SCIENCE ADVANCES 2022; 8:eabq5772. [PMID: 36516264 PMCID: PMC9750137 DOI: 10.1126/sciadv.abq5772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 11/08/2022] [Indexed: 06/17/2023]
Abstract
BK channels uniquely integrate voltage and calcium signaling in diverse cell types through allosteric activation of their K+-conducting pore by structurally distinct V and Ca2+ sensor domains. Here, we define mechanisms and interaction pathways that link V sensors to the pore by analyzing effects on allosteric coupling of point mutations in the context of Slo1 BK channel structure. A gating lever, mediated by S4/S5 segment interaction within the transmembrane domain, rotates to engage and stabilize the open conformation of the S6 inner pore helix upon V sensor activation. In addition, an indirect pathway, mediated by the carboxyl-terminal cytosolic domain (CTD) and C-linker that connects the CTD to S6, stabilizes the closed conformation when V sensors are at rest. Unexpectedly, this mechanism, which bypasses the covalent connections of C-linker to CTD and pore, also transduces Ca2+-dependent coupling in a manner that is completely nonadditive with voltage, analogous to the function of a digital logic (OR) gate.
Collapse
Affiliation(s)
- Liang Sun
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank T. Horrigan
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
18
|
Liu X, Tao J, Zhang S, Lan W, Yao Y, Wang C, Xue H, Ji Y, Li G, Cao C. Development of charybdotoxin Q18F variant as a selective peptide blocker of neuronal BK(α + β4) channel for the treatment of epileptic seizures. Protein Sci 2022; 31:e4506. [PMID: 36369672 PMCID: PMC9703589 DOI: 10.1002/pro.4506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/14/2022]
Abstract
Epilepsy is the results from the imbalance between inhibition and excitation in neural circuits, which is mainly treated by some chemical drugs with side effects. Gain-of-function of BK channels or knockout of its β4 subunit associates with spontaneous epilepsy. Currently, few reports were published about the efficacy of BK(α + β4) channel modulators in epilepsy prevention. Charybdotoxin is a non-specific inhibitor of BK and other K+ channels. Here, by nuclear magnetic resonance (NMR) and other biochemical techniques, we found that charybdotoxin might interact with the extracellular loop of human β4 subunit (i.e., hβ4-loop) of BK(α + β4) channel at a molar ratio 4:1 (hβ4-loop vs. charybdotoxin). Charybdotoxin enhanced its ability to prevent K+ current of BK(α + β4 H101Y) channel. The charybdotoxin Q18F variant selectively reduced the neuronal spiking frequency and increased interspike intervals of BK(α + β4) channel by π-π stacking interactions between its residue Phe18 and residue His101 of hβ4-loop. Moreover, intrahippocampal infusion of charybdotoxin Q18F variant significantly increased latency time of seizure, reduced seizure duration and seizure numbers on pentylenetetrazole-induced pre-sensitized rats, inhibited hippocampal hyperexcitability and c-Fos expression, and displayed neuroprotective effects on hippocampal neurons. These results implied that charybdotoxin Q18F variant could be potentially used for intractable epilepsy treatment by therapeutically targeting BK(α + β4) channel.
Collapse
Affiliation(s)
- Xinlian Liu
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of ScienceBeijingChina
| | - Jie Tao
- Department of Neurology and Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Biomembrane and BiopharmaceuticsShanghai UniversityShanghaiChina
| | - Shuzhang Zhang
- Institute of Biomembrane and BiopharmaceuticsShanghai UniversityShanghaiChina
| | - Wenxian Lan
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
| | - Yu Yao
- Institute of Biomembrane and BiopharmaceuticsShanghai UniversityShanghaiChina
| | - Chunxi Wang
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
| | - Hongjuan Xue
- National Facility for Protein Science in Shanghai, Zhangjiang LabShanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Yonghua Ji
- Institute of Biomembrane and BiopharmaceuticsShanghai UniversityShanghaiChina
| | - Guoyi Li
- Department of Neurology and Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chunyang Cao
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of ScienceBeijingChina
| |
Collapse
|
19
|
Zhang G, Xu X, Jia Z, Geng Y, Liang H, Shi J, Marras M, Abella C, Magleby KL, Silva JR, Chen J, Zou X, Cui J. An allosteric modulator activates BK channels by perturbing coupling between Ca 2+ binding and pore opening. Nat Commun 2022; 13:6784. [PMID: 36351900 PMCID: PMC9646747 DOI: 10.1038/s41467-022-34359-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022] Open
Abstract
BK type Ca2+-activated K+ channels activate in response to both voltage and Ca2+. The membrane-spanning voltage sensor domain (VSD) activation and Ca2+ binding to the cytosolic tail domain (CTD) open the pore across the membrane, but the mechanisms that couple VSD activation and Ca2+ binding to pore opening are not clear. Here we show that a compound, BC5, identified from in silico screening, interacts with the CTD-VSD interface and specifically modulates the Ca2+ dependent activation mechanism. BC5 activates the channel in the absence of Ca2+ binding but Ca2+ binding inhibits BC5 effects. Thus, BC5 perturbs a pathway that couples Ca2+ binding to pore opening to allosterically affect both, which is further supported by atomistic simulations and mutagenesis. The results suggest that the CTD-VSD interaction makes a major contribution to the mechanism of Ca2+ dependent activation and is an important site for allosteric agonists to modulate BK channel activation.
Collapse
Affiliation(s)
- Guohui Zhang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA
| | - Xianjin Xu
- Dalton Cardiovascular Research Center, University of Missouri - Columbia, Columbia, MO, USA.,Department of Physics and Astronomy, University of Missouri - Columbia, Columbia, MO, USA.,Department of Biochemistry, University of Missouri - Columbia, Columbia, MO, USA.,Institute for Data Science and Informatics, University of Missouri - Columbia, Columbia, MO, USA
| | - Zhiguang Jia
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA.,Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| | - Yanyan Geng
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hongwu Liang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA
| | - Martina Marras
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA
| | - Carlota Abella
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA
| | - Karl L Magleby
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan R Silva
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA.
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA. .,Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA.
| | - Xiaoqin Zou
- Dalton Cardiovascular Research Center, University of Missouri - Columbia, Columbia, MO, USA. .,Department of Physics and Astronomy, University of Missouri - Columbia, Columbia, MO, USA. .,Department of Biochemistry, University of Missouri - Columbia, Columbia, MO, USA. .,Institute for Data Science and Informatics, University of Missouri - Columbia, Columbia, MO, USA.
| | - Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
20
|
Srinivasan SR, Huang H, Chang WC, Nasburg JA, Nguyen HM, Strassmaier T, Wulff H, Shakkottai VG. Discovery of Novel Activators of Large-Conductance Calcium-Activated Potassium Channels for the Treatment of Cerebellar Ataxia. Mol Pharmacol 2022; 102:438-449. [PMID: 35489717 PMCID: PMC9341255 DOI: 10.1124/molpharm.121.000478] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Impaired cerebellar Purkinje neuron firing resulting from reduced expression of large-conductance calcium-activated potassium (BK) channels is a consistent feature in models of inherited neurodegenerative spinocerebellar ataxia (SCA). Restoring BK channel expression improves motor function and delays cerebellar degeneration, indicating that BK channels are an attractive therapeutic target. Current BK channel activators lack specificity and potency and are therefore poor templates for future drug development. We implemented an automated patch clamp platform for high-throughput drug discovery of BK channel activators using the Nanion SyncroPatch 384PE system. We screened over 15,000 compounds for their ability to increase BK channel current amplitude under conditions of lower intracellular calcium that is present in disease. We identified several novel BK channel activators that were then retested on the SyncroPatch 384PE to generate concentration-response curves (CRCs). Compounds with favorable CRCs were subsequently tested for their ability to improve irregular cerebellar Purkinje neuron spiking, characteristic of BK channel dysfunction in SCA1 mice. We identified a novel BK channel activator, 4-chloro-N-(5-chloro-2-cyanophenyl)-3-(trifluoromethyl)benzene-1-sulfonamide (herein renamed BK-20), that exhibited a more potent half-maximal activation of BK current (pAC50 = 4.64) than NS-1619 (pAC50 = 3.7) at a free internal calcium concentration of 270 nM in a heterologous expression system and improved spiking regularity in SCA1 Purkinje neurons. BK-20 had no activity on small-conductance calcium-activated potassium (SK)1-3 channels but interestingly was a potent blocker of the T-type calcium channel, Cav3.1 (IC50 = 1.05 μM). Our work describes both a novel compound for further drug development in disorders with irregular Purkinje spiking and a unique platform for drug discovery in degenerative ataxias. SIGNIFICANCE STATEMENT: Motor impairment associated with altered Purkinje cell spiking due to dysregulation of large-conductance calcium-activated potassium (BK) channel expression and function is a shared feature of disease in many degenerative ataxias. BK channel activators represent an outstanding therapeutic agent for ataxia. We have developed a high-throughput platform to screen for BK channel activators and identified a novel compound that can serve as a template for future drug development for the treatment of these disabling disorders.
Collapse
Affiliation(s)
- Sharan R Srinivasan
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Haoran Huang
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Wei-Chih Chang
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Joshua A Nasburg
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Hai M Nguyen
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Tim Strassmaier
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Heike Wulff
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Vikram G Shakkottai
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| |
Collapse
|
21
|
Zuccolini P, Gavazzo P, Pusch M. BK Channel in the Physiology and in the Cancer of Pancreatic Duct: Impact and Reliability of BK Openers. Front Pharmacol 2022; 13:906608. [PMID: 35685628 PMCID: PMC9171006 DOI: 10.3389/fphar.2022.906608] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
BK (KCa 1.1, Slo-1) is a K+ channel characterized by an allosteric regulation of the gating mechanism by Ca2+ binding and voltage, and a high unitary conductance. The channel is expressed in many different tissues, where it is involved in the regulation or the fine-tuning of many physiological processes. Among other organs, BK is expressed in the pancreatic duct, a part of the gland important for the correct ionic composition of the pancreatic juice. Unfortunately, the pancreatic duct is also the site where one of the deadliest cancer types, the pancreatic duct adenocarcinoma (PDAC), develops. In the past years, it has been reported that continuous exposure of cancer cells to BK openers can have a significant impact on cell viability as well as on the ability to proliferate and migrate. Here, we first summarize the main BK channel properties and its roles in pancreatic duct physiology. Then we focus on the potential role of BK as a pharmacological target in PDAC. Moreover, we discuss how results obtained when employing BK activators on cancer cells can, in some cases, be misleading.
Collapse
|
22
|
Sanghvi S, Szteyn K, Ponnalagu D, Sridharan D, Lam A, Hansra I, Chaudhury A, Majumdar U, Kohut AR, Gururaja Rao S, Khan M, Garg V, Singh H. Inhibition of BK Ca channels protects neonatal hearts against myocardial ischemia and reperfusion injury. Cell Death Dis 2022; 8:175. [PMID: 35393410 PMCID: PMC8989942 DOI: 10.1038/s41420-022-00980-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
Abstract
BKCa channels are large-conductance calcium and voltage-activated potassium channels that are heterogeneously expressed in a wide array of cells. Activation of BKCa channels present in mitochondria of adult ventricular cardiomyocytes is implicated in cardioprotection against ischemia-reperfusion (IR) injury. However, the BKCa channel’s activity has never been detected in the plasma membrane of adult ventricular cardiomyocytes. In this study, we report the presence of the BKCa channel in the plasma membrane and mitochondria of neonatal murine and rodent cardiomyocytes, which protects the heart on inhibition but not activation. Furthermore, K+ currents measured in neonatal cardiomyocyte (NCM) was sensitive to iberiotoxin (IbTx), suggesting the presence of BKCa channels in the plasma membrane. Neonatal hearts subjected to IR when post-conditioned with NS1619 during reoxygenation increased the myocardial infarction whereas IbTx reduced the infarct size. In agreement, isolated NCM also presented increased apoptosis on treatment with NS1619 during hypoxia and reoxygenation, whereas IbTx reduced TUNEL-positive cells. In NCMs, activation of BKCa channels increased the intracellular reactive oxygen species post HR injury. Electrophysiological characterization of NCMs indicated that NS1619 increased the beat period, field, and action potential duration, and decreased the conduction velocity and spike amplitude. In contrast, IbTx had no impact on the electrophysiological properties of NCMs. Taken together, our data established that inhibition of plasma membrane BKCa channels in the NCM protects neonatal heart/cardiomyocytes from IR injury. Furthermore, the functional disparity observed towards the cardioprotective activity of BKCa channels in adults compared to neonatal heart could be attributed to their differential localization.
Collapse
Affiliation(s)
- Shridhar Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Kalina Szteyn
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Divya Sridharan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Alexander Lam
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Inderjot Hansra
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ankur Chaudhury
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Uddalak Majumdar
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrew R Kohut
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA.,Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shubha Gururaja Rao
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, OH, USA
| | - Mahmood Khan
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Gonzalez-Perez V, Zhou Y, Ciorba MA, Lingle CJ. The LRRC family of BK channel regulatory subunits: potential roles in health and disease. J Physiol 2022; 600:1357-1371. [PMID: 35014034 PMCID: PMC8930516 DOI: 10.1113/jp281952] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
Large conductance K+ channels, termed BK channels, regulate a variety of cellular and physiological functions. Although universally activated by changes in voltage or [Ca2+ ]i , the threshold for BK channel activation varies among loci of expression, often arising from cell-specific regulatory subunits including a family of leucine rich repeat-containing (LRRC) γ subunits (LRRC26, LRRC52, LRRC55 and LRRC38). The 'founding' member of this family, LRRC26, was originally identified as a tumour suppressor in various cancers. An LRRC26 knockout (KO) mouse model recently revealed that LRRC26 is also highly expressed in secretory epithelial cells and partners with BK channels in the salivary gland and colonic goblet cells to promote sustained K+ fluxes likely essential for normal secretory function. To accomplish this, LRRC26 negatively shifts the range of BK channel activation such that channels contribute to K+ flux near typical epithelial cell resting conditions. In colon, the absence of LRRC26 increases vulnerability to colitis. LRRC26-containing BK channels are also likely important regulators of epithelial function in other loci, including airways, female reproductive tract and mammary gland. Based on an LRRC52 KO mouse model, LRRC52 regulation of large conductance K+ channels plays a role both in sperm function and in cochlear inner hair cells. Although our understanding of LRRC-containing BK channels remains rudimentary, KO mouse models may help define other organs in which LRRC-containing channels support normal function. A key topic for future work concerns identification of endogenous mechanisms, whether post-translational or via gene regulation, that may impact LRRC-dependent pathologies.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Yu Zhou
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew A Ciorba
- Department of Internal Medicine, Division of Gastroenterology, Washington University School of Medicine, St Louis, MO, USA
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
24
|
Sanghvi S, Szteyn K, Ponnalagu D, Sridharan D, Lam A, Hansra I, Chaudhury A, Majumdar U, Kohut AR, Rao SG, Khan M, Garg V, Singh H. Inhibition of BK Ca channels protects neonatal hearts against myocardial ischemia and reperfusion injury.. [DOI: 10.1101/2021.11.02.466585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
Abstract
AbstractBKCa channels are large-conductance calcium and voltage-activated potassium channels that are heterogeneously expressed in a wide array of cells. Activation of BKCa channels present in mitochondria of adult ventricular cardiomyocytes is implicated in cardioprotection against ischemia-reperfusion (IR) injury. However, the BKCa channel’s activity has never been detected in the plasma membrane of adult ventricular cardiomyocytes. In this study, we report the presence of the BKCa channel in the plasma membrane and mitochondria of neonatal murine and rodent cardiomyocytes which protects the heart on inhibition but not activation. Furthermore, K+ currents measured in neonatal cardiomyocyte (NCM) was sensitive to iberiotoxin (IbTx), suggesting the presence of BKCa channels in the plasma membrane. Neonatal hearts subjected to IR when post-conditioned with NS1619 during reoxygenation increased the myocardial infarction whereas IbTx reduced the infarct size. In agreement, isolated NCM also presented increased apoptosis on treatment with NS1619 during hypoxia and reoxygenation, whereas IbTx reduced TUNEL positive cells. In NCMs, activation of BKCa channels increased the intracellular reactive oxygen species post HR injury. Electrophysiological characterization of NCMs indicated that NS1619 increased the beat period, field, and action potential duration, and decreased the conduction velocity and spike amplitude. In contrast, IbTx had no impact on the electrophysiological properties of NCMs. Taken together, our data established that inhibition of plasma membrane BKCa channels in the NCM protects neonatal heart/cardiomyocytes from IR injury. Furthermore, the functional disparity observed towards the cardioprotective activity of BKCa channels in adults compared to neonatal heart could be attributed to their differential localization.
Collapse
|
25
|
Cui J. BK Channel Gating Mechanisms: Progresses Toward a Better Understanding of Variants Linked Neurological Diseases. Front Physiol 2021; 12:762175. [PMID: 34744799 PMCID: PMC8567085 DOI: 10.3389/fphys.2021.762175] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/01/2021] [Indexed: 12/21/2022] Open
Abstract
The large conductance Ca2+-activated potassium (BK) channel is activated by both membrane potential depolarization and intracellular Ca2+ with distinct mechanisms. Neural physiology is sensitive to the function of BK channels, which is shown by the discoveries of neurological disorders that are associated with BK channel mutations. This article reviews the molecular mechanisms of BK channel activation in response to voltage and Ca2+ binding, including the recent progress since the publication of the atomistic structure of the whole BK channel protein, and the neurological disorders associated with BK channel mutations. These results demonstrate the unique mechanisms of BK channel activation and that these mechanisms are important factors in linking BK channel mutations to neurological disorders.
Collapse
Affiliation(s)
- Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St. Louis, MO, United States
| |
Collapse
|
26
|
Kshatri A, Cerrada A, Gimeno R, Bartolomé-Martín D, Rojas P, Giraldez T. Differential regulation of BK channels by fragile X mental retardation protein. J Gen Physiol 2021; 152:151651. [PMID: 32275741 PMCID: PMC7266151 DOI: 10.1085/jgp.201912502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/12/2020] [Indexed: 01/28/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein prominently expressed in neurons. Missense mutations or complete loss of FMRP can potentially lead to fragile X syndrome, a common form of inherited intellectual disability. In addition to RNA regulation, FMRP was also proposed to modulate neuronal function by direct interaction with the large conductance Ca2+- and voltage-activated potassium channel (BK) β4 regulatory subunits (BKβ4). However, the molecular mechanisms underlying FMRP regulation of BK channels were not studied in detail. We have used electrophysiology and super-resolution stochastic optical reconstruction microscopy (STORM) to characterize the effects of FMRP on pore-forming BKα subunits, as well as the association with regulatory subunits BKβ4. Our data indicate that, in the absence of coexpressed β4, FMRP alters the steady-state properties of BKα channels by decreasing channel activation and deactivation rates. Analysis using the Horrigan-Aldrich model revealed alterations in the parameters associated with channel opening (L0) and voltage sensor activation (J0). Interestingly, FMRP also altered the biophysical properties of BKαβ4 channels favoring channel opening, although not as dramatically as BKα. STORM experiments revealed clustered multi-protein complexes, consistent with FMRP interacting not only to BKαβ4 but also to BKα. Lastly, we found that a partial loss-of-function mutation in FMRP (R138Q) counteracts many of its functional effects on BKα and BKαβ4 channels. In summary, our data show that FMRP modulates the function of both BKα and BKαβ4 channels.
Collapse
Affiliation(s)
- Aravind Kshatri
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alejandro Cerrada
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Roger Gimeno
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - David Bartolomé-Martín
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Teresa Giraldez
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|
27
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and remyelination. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
28
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and the preservation of myelin. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
29
|
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
|
30
|
Sek A, Kampa RP, Kulawiak B, Szewczyk A, Bednarczyk P. Identification of the Large-Conductance Ca 2+-Regulated Potassium Channel in Mitochondria of Human Bronchial Epithelial Cells. Molecules 2021; 26:molecules26113233. [PMID: 34072205 PMCID: PMC8199365 DOI: 10.3390/molecules26113233] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria play a key role in energy metabolism within the cell. Potassium channels such as ATP-sensitive, voltage-gated or large-conductance Ca2+-regulated channels have been described in the inner mitochondrial membrane. Several hypotheses have been proposed to describe the important roles of mitochondrial potassium channels in cell survival and death pathways. In the current study, we identified two populations of mitochondrial large-conductance Ca2+-regulated potassium (mitoBKCa) channels in human bronchial epithelial (HBE) cells. The biophysical properties of the channels were characterized using the patch-clamp technique. We observed the activity of the channel with a mean conductance close to 285 pS in symmetric 150/150 mM KCl solution. Channel activity was increased upon application of the potassium channel opener NS11021 in the micromolar concentration range. The channel activity was completely inhibited by 1 µM paxilline and 300 nM iberiotoxin, selective inhibitors of the BKCa channels. Based on calcium and iberiotoxin modulation, we suggest that the C-terminus of the protein is localized to the mitochondrial matrix. Additionally, using RT-PCR, we confirmed the presence of α pore-forming (Slo1) and auxiliary β3-β4 subunits of BKCa channel in HBE cells. Western blot analysis of cellular fractions confirmed the mitochondrial localization of α pore-forming and predominately β3 subunits. Additionally, the regulation of oxygen consumption and membrane potential of human bronchial epithelial mitochondria in the presence of the potassium channel opener NS11021 and inhibitor paxilline were also studied. In summary, for the first time, the electrophysiological and functional properties of the mitoBKCa channel in a bronchial epithelial cell line were described.
Collapse
Affiliation(s)
- Aleksandra Sek
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | - Rafal P. Kampa
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences—SGGW, 02-776 Warsaw, Poland
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences—SGGW, 02-776 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-593-8620
| |
Collapse
|
31
|
Role of Airway Smooth Muscle in Inflammation Related to Asthma and COPD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:139-172. [PMID: 33788192 DOI: 10.1007/978-3-030-63046-1_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Airway smooth muscle contributes to both contractility and inflammation in the pathophysiology of asthma and COPD. Airway smooth muscle cells can change the degree of a variety of functions, including contraction, proliferation, migration, and the secretion of inflammatory mediators (phenotype plasticity). Airflow limitation, airway hyperresponsiveness, β2-adrenergic desensitization, and airway remodeling, which are fundamental characteristic features of these diseases, are caused by phenotype changes in airway smooth muscle cells. Alterations between contractile and hyper-contractile, synthetic/proliferative phenotypes result from Ca2+ dynamics and Ca2+ sensitization. Modulation of Ca2+ dynamics through the large-conductance Ca2+-activated K+ channel/L-type voltage-dependent Ca2+ channel linkage and of Ca2+ sensitization through the RhoA/Rho-kinase pathway contributes not only to alterations in the contractile phenotype involved in airflow limitation, airway hyperresponsiveness, and β2-adrenergic desensitization but also to alteration of the synthetic/proliferative phenotype involved in airway remodeling. These Ca2+ signal pathways are also associated with synergistic effects due to allosteric modulation between β2-adrenergic agonists and muscarinic antagonists. Therefore, airway smooth muscle may be a target tissue in the therapy for these diseases. Moreover, the phenotype changing in airway smooth muscle cells with focuses on Ca2+ signaling may provide novel strategies for research and development of effective remedies against both bronchoconstriction and inflammation.
Collapse
|
32
|
Abstract
Potassium channels are the most diverse and ubiquitous family of ion channels found in cells. The Ca2+ and voltage gated members form a subfamily that play a variety of roles in both excitable and non-excitable cells and are further classified on the basis of their single channel conductance to form the small conductance (SK), intermediate conductance (IK) and big conductance (BK) K+ channels.In this chapter, we will focus on the mechanisms underlying the gating of BK channels, whose function is modified in different tissues by different splice variants as well as the expanding array of regulatory accessory subunits including β, γ and LINGO subunits. We will examine how BK channels are modified by these regulatory subunits and describe how the channel gating is altered by voltage and Ca2+ whilst setting this in context with the recently published structures of the BK channel. Finally, we will discuss how BK and other calcium-activated channels are modulated by novel ion channel modulators and describe some of the challenges associated with trying to develop compounds with sufficient efficacy, potency and selectivity to be of therapeutic benefit.
Collapse
|
33
|
Age-related hearing loss pertaining to potassium ion channels in the cochlea and auditory pathway. Pflugers Arch 2020; 473:823-840. [PMID: 33336302 PMCID: PMC8076138 DOI: 10.1007/s00424-020-02496-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/27/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly and constitutes the third highest risk factor for dementia. Lifetime noise exposure, genetic predispositions for degeneration, and metabolic stress are assumed to be the major causes of ARHL. Both noise-induced and hereditary progressive hearing have been linked to decreased cell surface expression and impaired conductance of the potassium ion channel KV7.4 (KCNQ4) in outer hair cells, inspiring future therapies to maintain or prevent the decline of potassium ion channel surface expression to reduce ARHL. In concert with KV7.4 in outer hair cells, KV7.1 (KCNQ1) in the stria vascularis, calcium-activated potassium channels BK (KCNMA1) and SK2 (KCNN2) in hair cells and efferent fiber synapses, and KV3.1 (KCNC1) in the spiral ganglia and ascending auditory circuits share an upregulated expression or subcellular targeting during final differentiation at hearing onset. They also share a distinctive fragility for noise exposure and age-dependent shortfalls in energy supply required for sustained surface expression. Here, we review and discuss the possible contribution of select potassium ion channels in the cochlea and auditory pathway to ARHL. We postulate genes, proteins, or modulators that contribute to sustained ion currents or proper surface expressions of potassium channels under challenging conditions as key for future therapies of ARHL.
Collapse
|
34
|
Mi R, Rabbi MH, Sun Y, Li X, Ma S, Wen Z, Meng N, Li Y, Du X, Li S. Enhanced protein phosphorylation in Apostichopus japonicus intestine triggered by tussah immunoreactive substances might be involved in the regulation of immune-related signaling pathways. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 37:100757. [PMID: 33197859 DOI: 10.1016/j.cbd.2020.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 11/28/2022]
Abstract
The sea cucumber Apostichopus japonicus is an economically important species owing to its high nutritive and medicinal value. In order to avoid the pollution resulting from the overuse of antibiotics in A. japonicus aquaculture, various immunostimulants have been used as an alternative to improve the efficiency of A. japonicus farming. Our previous proteomic investigation has shown that several proteins participating in the immune-related physiology of A. japonicus were differentially expressed in the intestinal tissue in response to tussah immunoreactive substances (TIS). This study further explored the immunostimulation mechanism of TIS in A. japonicus. Phosphoproteomics technology was used to investigate the effect of TIS on protein phosphorylation in the intestine of A. japonicus following feeding with a TIS-supplemented diet. A total of 213 unique phosphoproteins were detected from 225 unique phosphopeptides. KEGG pathway analysis showed that majority of the phosphoproteins are involved in endocytosis, carbon metabolism and spliceosome functional group. Sixteen of the phosphoproteins exhibited differential phosphorylation in response to TIS and 12 of these were found to associate with biological functions. Of these 12 phosphoproteins, eight exhibited enhanced phosphorylation while four displayed reduced phosphorylation. These 12 proteins were further analyzed and all were found to play a role in regulating some aspects of the immune system and the growth of sea cucumbers, especially in phagocytosis, energy metabolism and disease resistance. The findings of this study could therefore shed new light on the immune pathways of sea cucumber that are affected by TIS. This could help us to better understand the underlying mechanism linked to the immunoenhancement of A. japonicus in response to TIS, one that is associated with the change in protein phosphorylation.
Collapse
Affiliation(s)
- Rui Mi
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Md Hasim Rabbi
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116024, PR China
| | - Yongxin Sun
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China.
| | - Xuejun Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Shuhui Ma
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Zhixin Wen
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Nan Meng
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Yajie Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Xingfan Du
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| | - Shuying Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian 116024, PR China
| |
Collapse
|
35
|
Zhang G, Gibson RA, McDonald M, Liang P, Kang PW, Shi J, Yang H, Cui J, Mikati MA. A Gain-of-Function Mutation in KCNMA1 Causes Dystonia Spells Controlled With Stimulant Therapy. Mov Disord 2020; 35:1868-1873. [PMID: 32633875 DOI: 10.1002/mds.28138] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The mutations of KCNMA1 BK-type K+ channel have been identified in patients with various movement disorders. The underlying pathophysiology and corresponding therapeutics are lacking. OBJECTIVES To report our clinical and biophysical characterizations of a novel de novo KCNMA1 variant, as well as an effective therapy for the patient's dystonia-atonia spells. METHODS Combination of phenotypic characterization, therapy, and biophysical characterization of the patient and her mutation. RESULTS The patient had >100 dystonia-atonia spells per day with mild cerebellar atrophy. She also had autism spectrum disorder, intellectual disability, and attention deficit hyperactivity disorder. Whole-exome sequencing identified a heterozygous de novo BK N536H mutation. Our biophysical characterization demonstrates that N536H is a gain-of-function mutation with markedly enhanced voltage-dependent activation. Remarkably, administration of dextroamphetamine completely suppressed the dystonia-atonia spells. CONCLUSIONS BK N536H is a gain-of-function that causes dystonia and other neurological symptoms. Our stimulant therapy opens a new avenue to mitigate KCNMA1-linked movement disorders. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Guohui Zhang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rebecca A Gibson
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Marie McDonald
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
| | - Po Wei Kang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mohamad A Mikati
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
36
|
Yazdani M, Zhang G, Jia Z, Shi J, Cui J, Chen J. Aromatic interactions with membrane modulate human BK channel activation. eLife 2020; 9:55571. [PMID: 32597752 PMCID: PMC7371421 DOI: 10.7554/elife.55571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/28/2020] [Indexed: 12/22/2022] Open
Abstract
Large-conductance potassium (BK) channels are transmembrane (TM) proteins that can be synergistically and independently activated by membrane voltage and intracellular Ca2+. The only covalent connection between the cytosolic Ca2+ sensing domain and the TM pore and voltage sensing domains is a 15-residue ‘C-linker’. To determine the linker’s role in human BK activation, we designed a series of linker sequence scrambling mutants to suppress potential complex interplay of specific interactions with the rest of the protein. The results revealed a surprising sensitivity of BK activation to the linker sequence. Combining atomistic simulations and further mutagenesis experiments, we demonstrated that nonspecific interactions of the linker with membrane alone could directly modulate BK activation. The C-linker thus plays more direct roles in mediating allosteric coupling between BK domains than previously assumed. Our results suggest that covalent linkers could directly modulate TM protein function and should be considered an integral component of the sensing apparatus.
Collapse
Affiliation(s)
- Mahdieh Yazdani
- Department of Chemistry, University of Massachusetts, Amherst, United States
| | - Guohui Zhang
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, United States
| | - Zhiguang Jia
- Department of Chemistry, University of Massachusetts, Amherst, United States
| | - Jingyi Shi
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, United States
| | - Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Cardiac Bioelectricity and Arrhythmia Center, Washington University, St Louis, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, United States.,Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, United States
| |
Collapse
|
37
|
Coupling of Ca 2+ and voltage activation in BK channels through the αB helix/voltage sensor interface. Proc Natl Acad Sci U S A 2020; 117:14512-14521. [PMID: 32513714 DOI: 10.1073/pnas.1908183117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Large-conductance Ca2+ and voltage-activated K+ (BK) channels control membrane excitability in many cell types. BK channels are tetrameric. Each subunit is composed of a voltage sensor domain (VSD), a central pore-gate domain, and a large cytoplasmic domain (CTD) that contains the Ca2+ sensors. While it is known that BK channels are activated by voltage and Ca2+, and that voltage and Ca2+ activations interact, less is known about the mechanisms involved. We explore here these mechanisms by examining the gating contribution of an interface formed between the VSDs and the αB helices located at the top of the CTDs. Proline mutations in the αB helix greatly decreased voltage activation while having negligible effects on gating currents. Analysis with the Horrigan, Cui, and Aldrich model indicated a decreased coupling between voltage sensors and pore gate. Proline mutations decreased Ca2+ activation for both Ca2+ bowl and RCK1 Ca2+ sites, suggesting that both high-affinity Ca2+ sites transduce their effect, at least in part, through the αB helix. Mg2+ activation also decreased. The crystal structure of the CTD with proline mutation L390P showed a flattening of the first helical turn in the αB helix compared to wild type, without other notable differences in the CTD, indicating that structural changes from the mutation were confined to the αB helix. These findings indicate that an intact αB helix/VSD interface is required for effective coupling of Ca2+ binding and voltage depolarization to pore opening and that shared Ca2+ and voltage transduction pathways involving the αB helix may be involved.
Collapse
|
38
|
Abstract
Ca2+- and voltage-gated K+ channels of large conductance (BK channels) are expressed in a diverse variety of both excitable and inexcitable cells, with functional properties presumably uniquely calibrated for the cells in which they are found. Although some diversity in BK channel function, localization, and regulation apparently arises from cell-specific alternative splice variants of the single pore-forming α subunit ( KCa1.1, Kcnma1, Slo1) gene, two families of regulatory subunits, β and γ, define BK channels that span a diverse range of functional properties. We are just beginning to unravel the cell-specific, physiological roles served by BK channels of different subunit composition.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
39
|
Scruggs AM, Grabauskas G, Huang SK. The Role of KCNMB1 and BK Channels in Myofibroblast Differentiation and Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2020; 62:191-203. [PMID: 31486669 DOI: 10.1165/rcmb.2019-0163oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The differentiation of fibroblasts into myofibroblasts is critical for the development of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF). Previously, we demonstrated that fibroblasts from patients with IPF exhibit changes in DNA methylation across the genome that contribute to a profibrotic phenotype. One of the top differentially methylated genes identified in our previous study was KCNMB1, which codes for the β subunit of the large-conductance potassium (BK, also known as MaxiK or KCa1.1) channel. Here, we examined how the expression of KCNMB1 differed between IPF fibroblasts and normal cells, and how BK channels affected myofibroblast differentiation. Fibroblasts from patients with IPF exhibited increased expression of KCNMB1, which corresponded to increased DNA methylation within the gene body. Patch-clamp experiments demonstrated that IPF fibroblasts had increased BK channel activity. Knockdown of KCNMB1 attenuated the ability of fibroblasts to contract collagen gels, and this was associated with a loss of α-smooth muscle actin (SMA) expression. Pharmacologic activation of BK channels stimulated α-SMA expression, whereas BK channel inhibitors blocked the upregulation of α-SMA. The ability of BK channels to enhance α-SMA expression was dependent on intracellular calcium, as activation of BK channels resulted in increased levels of intracellular calcium and the effects of BK agonists were abolished when calcium was removed. Together, our findings demonstrate that epigenetic upregulation of KCNMB1 contributes to increased BK channel activity in IPF fibroblasts, and identify a newfound role for BK channels in myofibroblast differentiation.
Collapse
Affiliation(s)
| | - Gintautas Grabauskas
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
40
|
Trombetta-Lima M, Krabbendam IE, Dolga AM. Calcium-activated potassium channels: implications for aging and age-related neurodegeneration. Int J Biochem Cell Biol 2020; 123:105748. [PMID: 32353429 DOI: 10.1016/j.biocel.2020.105748] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Population aging, as well as the handling of age-associated diseases, is a worldwide increasing concern. Among them, Alzheimer's disease stands out as the major cause of dementia culminating in full dependence on other people for basic functions. However, despite numerous efforts, in the last decades, there was no new approved therapeutic drug for the treatment of the disease. Calcium-activated potassium channels have emerged as a potential tool for neuronal protection by modulating intracellular calcium signaling. Their subcellular localization is determinant of their functional effects. When located on the plasma membrane of neuronal cells, they can modulate synaptic function, while their activation at the inner mitochondrial membrane has a neuroprotective potential via the attenuation of mitochondrial reactive oxygen species in conditions of oxidative stress. Here we review the dual role of these channels in the aging phenotype and Alzheimer's disease pathology and discuss their potential use as a therapeutic tool.
Collapse
Affiliation(s)
- Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands; Medical School, Neurology Department, University of São Paulo (USP), 01246903 São Paulo, Brazil
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
41
|
Yeste M, Llavanera M, Pérez G, Scornik F, Puig-Parri J, Brugada R, Bonet S, Pinart E. Elucidating the Role of K + Channels during In Vitro Capacitation of Boar Spermatozoa: Do SLO1 Channels Play a Crucial Role? Int J Mol Sci 2019; 20:E6330. [PMID: 31847486 PMCID: PMC6940911 DOI: 10.3390/ijms20246330] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/16/2023] Open
Abstract
This study sought to identify and localize SLO1 channels in boar spermatozoa by immunoblotting and immunofluorescence, and to determine their physiological role during in vitro sperm capacitation. Sperm samples from 14 boars were incubated in a capacitation medium for 300 min in the presence of paxilline (PAX), a specific SLO1-channel blocker, added either at 0 min or after 240 min of incubation. Negative controls were incubated in capacitation medium, and positive controls in capacitation medium plus tetraethyl ammonium (TEA), a general K+-channel blocker, also added at 0 min or after 240 min of incubation. In all samples, acrosome exocytosis was triggered with progesterone after 240 min of incubation. Sperm motility and kinematics, integrity of plasma and acrosome membranes, membrane lipid disorder, intracellular calcium levels and acrosin activity were evaluated after 0, 60, 120, 180, 240, 250, 270 and 300 min of incubation. In boar spermatozoa, SLO1 channels were found to have 80 kDa and be localized in the anterior postacrosomal region and the mid and principal piece of the tail; their specific blockage through PAX resulted in altered calcium levels and acrosome exocytosis. As expected, TEA blocker impaired in vitro sperm capacitation, by altering sperm motility and kinematics and calcium levels. In conclusion, SLO1 channels are crucial for the acrosome exocytosis induced by progesterone in in vitro capacitated boar spermatozoa.
Collapse
Affiliation(s)
- Marc Yeste
- Unit of Cell Biology, Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Faculty of Sciences, Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (J.P.-P.); (S.B.)
| | - Marc Llavanera
- Unit of Cell Biology, Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Faculty of Sciences, Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (J.P.-P.); (S.B.)
| | - Guillermo Pérez
- Department of Medical Sciences, Faculty of Medicine, University of Girona, E-17003 Girona, Spain; (G.P.); (F.S.); (R.B.)
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), E-17190 Girona, Spain
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), E-28029 Madrid, Spain
| | - Fabiana Scornik
- Department of Medical Sciences, Faculty of Medicine, University of Girona, E-17003 Girona, Spain; (G.P.); (F.S.); (R.B.)
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), E-17190 Girona, Spain
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), E-28029 Madrid, Spain
| | - Josep Puig-Parri
- Unit of Cell Biology, Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Faculty of Sciences, Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (J.P.-P.); (S.B.)
| | - Ramon Brugada
- Department of Medical Sciences, Faculty of Medicine, University of Girona, E-17003 Girona, Spain; (G.P.); (F.S.); (R.B.)
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), E-17190 Girona, Spain
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), E-28029 Madrid, Spain
- Cardiology Service, Hospital Josep Trueta, E-17003 Girona, Spain
| | - Sergi Bonet
- Unit of Cell Biology, Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Faculty of Sciences, Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (J.P.-P.); (S.B.)
| | - Elisabeth Pinart
- Unit of Cell Biology, Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Faculty of Sciences, Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (J.P.-P.); (S.B.)
| |
Collapse
|
42
|
Liu X, Tao J, Zhang S, Lan W, Wang C, Ji Y, Cao C. Selective Blockade of Neuronal BK (α + β4) Channels Preventing Epileptic Seizure. J Med Chem 2019; 63:216-230. [DOI: 10.1021/acs.jmedchem.9b01241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xinlian Liu
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
- University of Chinese Academy of Science, No. 19A, Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Jie Tao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Putuo District, Shanghai 200062, China
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, 99 Shangda Road,
BaoShan District, Shanghai 200444, China
| | - Shuzhang Zhang
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, 99 Shangda Road,
BaoShan District, Shanghai 200444, China
| | - Wenxian Lan
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Chunxi Wang
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Yonghua Ji
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, 99 Shangda Road,
BaoShan District, Shanghai 200444, China
- Xinhua Hospital (Chongming) Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai Chongming Xinhua Translational Medical Institute for Cancer Pain, 25 Nanmen Port Street, Chongming Branch, Shanghai 202150, China
| | - Chunyang Cao
- State Key Laboratory of Bioorganic and Natural Product Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
- University of Chinese Academy of Science, No. 19A, Yuquan Road, Shijingshan District, Beijing 100049, China
- Institute of Drug Discovery Technology, Ningbo University, No 818 Fenghua Road, Ningbo, Zhejiang 313211, China
| |
Collapse
|
43
|
Tao X, MacKinnon R. Molecular structures of the human Slo1 K + channel in complex with β4. eLife 2019; 8:51409. [PMID: 31815672 PMCID: PMC6934384 DOI: 10.7554/elife.51409] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022] Open
Abstract
Slo1 is a Ca2+- and voltage-activated K+ channel that underlies skeletal and smooth muscle contraction, audition, hormone secretion and neurotransmitter release. In mammals, Slo1 is regulated by auxiliary proteins that confer tissue-specific gating and pharmacological properties. This study presents cryo-EM structures of Slo1 in complex with the auxiliary protein, β4. Four β4, each containing two transmembrane helices, encircle Slo1, contacting it through helical interactions inside the membrane. On the extracellular side, β4 forms a tetrameric crown over the pore. Structures with high and low Ca2+ concentrations show that identical gating conformations occur in the absence and presence of β4, implying that β4 serves to modulate the relative stabilities of 'pre-existing' conformations rather than creating new ones. The effects of β4 on scorpion toxin inhibition kinetics are explained by the crown, which constrains access but does not prevent binding.
Collapse
Affiliation(s)
- Xiao Tao
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, New York, United States
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, New York, United States
| |
Collapse
|
44
|
Plante AE, Lai MH, Lu J, Meredith AL. Effects of Single Nucleotide Polymorphisms in Human KCNMA1 on BK Current Properties. Front Mol Neurosci 2019; 12:285. [PMID: 31849601 PMCID: PMC6901604 DOI: 10.3389/fnmol.2019.00285] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
BK Ca2+-activated K+ channels are important regulators of membrane excitability. Multiple regulatory mechanisms tailor BK current properties across tissues, such as alternative splicing, posttranslational modifications, and auxiliary subunits. Another potential mechanism for modulating BK channel activity is genetic variation due to single nucleotide polymorphisms (SNPs). The gene encoding the human BK α subunit, KCNMA1, contains hundreds of SNPs. However, the variation in BK channel activity due to SNPs is not well studied. Here, we screened the effects of four SNPs (A138V, C495G, N599D, and R800W) on BK currents in HEK293T cells, selected based on predicted protein pathogenicity or disease linkage. We found that the SNPs C495G and R800W had the largest effects on BK currents, affecting the conductance-voltage relationship across multiple Ca2+ conditions in the context of two BK channel splice variants. In symmetrical K+, C495G shifted the V1/2 to more hyperpolarized potentials (by -15 to -20 mV) and accelerated activation, indicating C495G confers some gain-of-function properties. R800W shifted the V1/2 to more depolarized potentials (+15 to +35 mV) and slowed activation, conferring loss-of-function properties. Moreover, the C495G and R800W effects on current properties were found to persist with posttranslational modifications. In contrast, A138V and N599D had smaller and more variable effects on current properties. Neither application of alkaline phosphatase to patches, which results in increased BK channel activity attributed to channel dephosphorylation, nor bidirectional redox modulations completely abrogated SNP effects on BK currents. Lastly, in physiological K+, C495G increased the amplitude of action potential (AP)-evoked BK currents, while R800W had a more limited effect. However, the introduction of R800W in parallel with the epilepsy-linked mutation D434G (D434G/R800W) decreased the amplitude of AP-evoked BK currents compared with D434G alone. These results suggest that in a physiological context, C495G could increase BK activation, while the effects of the loss-of-function SNP R800W could oppose the gain-of-function effects of an epilepsy-linked mutation. Together, these results implicate naturally occurring human genetic variation as a potential modifier of BK channel activity across a variety of conditions.
Collapse
Affiliation(s)
| | | | | | - Andrea L. Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
45
|
Kulik Y, Jones R, Moughamian AJ, Whippen J, Davis GW. Dual separable feedback systems govern firing rate homeostasis. eLife 2019; 8:45717. [PMID: 30973325 PMCID: PMC6491091 DOI: 10.7554/elife.45717] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/10/2019] [Indexed: 12/02/2022] Open
Abstract
Firing rate homeostasis (FRH) stabilizes neural activity. A pervasive and intuitive theory argues that a single variable, calcium, is detected and stabilized through regulatory feedback. A prediction is that ion channel gene mutations with equivalent effects on neuronal excitability should invoke the same homeostatic response. In agreement, we demonstrate robust FRH following either elimination of Kv4/Shal protein or elimination of the Kv4/Shal conductance. However, the underlying homeostatic signaling mechanisms are distinct. Eliminating Shal protein invokes Krüppel-dependent rebalancing of ion channel gene expression including enhanced slo, Shab, and Shaker. By contrast, expression of these genes remains unchanged in animals harboring a CRISPR-engineered, Shal pore-blocking mutation where compensation is achieved by enhanced IKDR. These different homeostatic processes have distinct effects on homeostatic synaptic plasticity and animal behavior. We propose that FRH includes mechanisms of proteostatic feedback that act in parallel with activity-driven feedback, with implications for the pathophysiology of human channelopathies.
Collapse
Affiliation(s)
- Yelena Kulik
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Ryan Jones
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Armen J Moughamian
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Jenna Whippen
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
46
|
Abstract
Ca2+- and voltage-gated K+ channels of large conductance (BK channels) are expressed in a diverse variety of both excitable and inexcitable cells, with functional properties presumably uniquely calibrated for the cells in which they are found. Although some diversity in BK channel function, localization, and regulation apparently arises from cell-specific alternative splice variants of the single pore-forming α subunit ( KCa1.1, Kcnma1, Slo1) gene, two families of regulatory subunits, β and γ, define BK channels that span a diverse range of functional properties. We are just beginning to unravel the cell-specific, physiological roles served by BK channels of different subunit composition.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
47
|
Regulatory γ1 subunits defy symmetry in functional modulation of BK channels. Proc Natl Acad Sci U S A 2018; 115:9923-9928. [PMID: 30224470 DOI: 10.1073/pnas.1804560115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Structural symmetry is a hallmark of homomeric ion channels. Nonobligatory regulatory proteins can also critically define the precise functional role of such channels. For instance, the pore-forming subunit of the large conductance voltage and calcium-activated potassium (BK, Slo1, or KCa1.1) channels encoded by a single KCa1.1 gene assembles in a fourfold symmetric fashion. Functional diversity arises from two families of regulatory subunits, β and γ, which help define the range of voltages over which BK channels in a given cell are activated, thereby defining physiological roles. A BK channel can contain zero to four β subunits per channel, with each β subunit incrementally influencing channel gating behavior, consistent with symmetry expectations. In contrast, a γ1 subunit (or single type of γ1 subunit complex) produces a functionally all-or-none effect, but the underlying stoichiometry of γ1 assembly and function remains unknown. Here we utilize two distinct and independent methods, a Forster resonance energy transfer-based optical approach and a functional reporter in single-channel recordings, to reveal that a BK channel can contain up to four γ1 subunits, but a single γ1 subunit suffices to induce the full gating shift. This requires that the asymmetric association of a single regulatory protein can act in a highly concerted fashion to allosterically influence conformational equilibria in an otherwise symmetric K+ channel.
Collapse
|
48
|
Rajendran VM, Sandle GI. Colonic Potassium Absorption and Secretion in Health and Disease. Compr Physiol 2018; 8:1513-1536. [PMID: 30215859 PMCID: PMC9769410 DOI: 10.1002/cphy.c170030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The colon has large capacities for K+ absorption and K+ secretion, but its role in maintaining K+ homeostasis is often overlooked. For many years, passive diffusion and/or solvent drag were thought to be the primary mechanisms for K+ absorption in human and animal colon. However, it is now clear that apical H+ ,K+ -ATPase, in coordination with basolateral K+ -Cl- cotransport and/or K+ and Cl- channels operating in parallel, mediate electroneutral K+ absorption in animal colon. We now know that K+ absorption in rat colon reflects ouabain-sensitive and ouabain-insensitive apical H+ ,K+ -ATPase activities. Ouabain-insensitive and ouabain-sensitive H+ ,K+ -ATPases are localized in surface and crypt cells, respectively. Colonic H+ ,K+ -ATPase consists of α- (HKCα ) and β- (HKCβ ) subunits which, when coexpressed, exhibit ouabain-insensitive H+ ,K+ -ATPase activity in HEK293 cells, while HKCα coexpressed with the gastric β-subunit exhibits ouabain-sensitive H+ ,K+ -ATPase activity in Xenopus oocytes. Aldosterone enhances apical H+ ,K+ -ATPase activity, HKCα specific mRNA and protein expression, and K+ absorption. Active K+ secretion, on the other hand, is mediated by apical K+ channels operating in a coordinated way with the basolateral Na+ -K+ -2Cl- cotransporter. Both Ca2+ -activated intermediate conductance K+ (IK) and large conductance K+ (BK) channels are located in the apical membrane of colonic epithelia. IK channel-mediated K+ efflux provides the driving force for Cl- secretion, while BK channels mediate active (e.g., cAMP-activated) K+ secretion. BK channel expression and activity are increased in patients with end-stage renal disease and ulcerative colitis. This review summarizes the role of apical H+ ,K+ -ATPase in K+ absorption, and apical BK channel function in K+ secretion in health and disease. © 2018 American Physiological Society. Compr Physiol 8:1513-1536, 2018.
Collapse
Affiliation(s)
| | - Geoffrey I. Sandle
- Leeds Institute of Biomedical and Clinical Sciences, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
49
|
Zhuang Z, Xiao J, Chen X, Hu X, Li R, Chen S, Feng X, Shen S, Ma HP, Zhuang J, Cai H. G protein pathway suppressor 2 enhanced the renal large-conductance Ca 2+-activated potassium channel expression via inhibiting ERK1/2 signaling pathway. Am J Physiol Renal Physiol 2018; 315:F503-F511. [PMID: 29767559 DOI: 10.1152/ajprenal.00041.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
G protein pathway suppressor 2 (GPS2) is a multifunctional protein and transcriptional regulation factor that is involved in the G protein MAPK signaling pathway. It has been shown that the MAPK signaling pathway plays an important role in the regulation of renal large-conductance Ca2+-activated potassium (BK) channels. In this study, we investigated the effects of GPS2 on BK channel activity and protein expression. In human embryonic kidney (HEK) BK stably expressing cells transfected with either GPS2 or its vector control, a single-cell recording showed that GPS2 significantly increased BK channel activity ( NPo), increasing BK open probability ( Po), and channel number ( N) compared with the control. In Cos-7 cells and HEK 293 T cells, GPS2 overexpression significantly enhanced the total protein expression of BK in a dose-dependent manner. Knockdown of GPS2 expression significantly decreased BK protein expression, while increasing ERK1/2 phosphorylation. Knockdown of ERK1/2 expression reversed the GPS2 siRNA-mediated inhibition of BK protein expression in Cos-7 cells. Pretreatments of Cos-7 cells with either the lysosomal inhibitor bafilomycin A1 or the proteasomal inhibitor MG132 partially reversed the inhibitory effects of GPS2 siRNA on BK protein expression. In addition, feeding a high-potassium diet significantly increased both GPS2 and BK protein abundance in mice. These data suggest that GPS2 enhances BK channel activity and its protein expression by reducing ERK1/2 signaling-mediated degradation of the channel.
Collapse
Affiliation(s)
- Zhizhi Zhuang
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China.,Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Jia Xiao
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia.,Xiangya Hospital, Central South University, Hunan, China
| | - Xinxin Chen
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China.,Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Xiaohan Hu
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China
| | - Ruidian Li
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China
| | - Shan Chen
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Xiuyan Feng
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia.,Xiangya Hospital, Central South University, Hunan, China
| | - Saier Shen
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Jieqiu Zhuang
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China
| | - Hui Cai
- Renal Division, the Second Affiliated Hospital, Wenzhou Medical University , Zhejiang , China.,Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia
| |
Collapse
|
50
|
Kshatri AS, Gonzalez-Hernandez A, Giraldez T. Physiological Roles and Therapeutic Potential of Ca 2+ Activated Potassium Channels in the Nervous System. Front Mol Neurosci 2018; 11:258. [PMID: 30104956 PMCID: PMC6077210 DOI: 10.3389/fnmol.2018.00258] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Within the potassium ion channel family, calcium activated potassium (KCa) channels are unique in their ability to couple intracellular Ca2+ signals to membrane potential variations. KCa channels are diversely distributed throughout the central nervous system and play fundamental roles ranging from regulating neuronal excitability to controlling neurotransmitter release. The physiological versatility of KCa channels is enhanced by alternative splicing and co-assembly with auxiliary subunits, leading to fundamental differences in distribution, subunit composition and pharmacological profiles. Thus, understanding specific KCa channels’ mechanisms in neuronal function is challenging. Based on their single channel conductance, KCa channels are divided into three subtypes: small (SK, 4–14 pS), intermediate (IK, 32–39 pS) and big potassium (BK, 200–300 pS) channels. This review describes the biophysical characteristics of these KCa channels, as well as their physiological roles and pathological implications. In addition, we also discuss the current pharmacological strategies and challenges to target KCa channels for the treatment of various neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Aravind S Kshatri
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alberto Gonzalez-Hernandez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Teresa Giraldez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|