1
|
Moldenhauer HJ, Tammen K, Meredith AL. Structural mapping of patient-associated KCNMA1 gene variants. Biophys J 2024; 123:1984-2000. [PMID: 38042986 PMCID: PMC11309989 DOI: 10.1016/j.bpj.2023.11.3404] [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: 09/08/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/04/2023] Open
Abstract
KCNMA1-linked channelopathy is a neurological disorder characterized by seizures, motor abnormalities, and neurodevelopmental disabilities. The disease mechanisms are predicted to result from alterations in KCNMA1-encoded BK K+ channel activity; however, only a subset of the patient-associated variants have been functionally studied. The localization of these variants within the tertiary structure or evaluation by pathogenicity algorithms has not been systematically assessed. In this study, 82 nonsynonymous patient-associated KCNMA1 variants were mapped within the BK channel protein. Fifty-three variants localized within cryoelectron microscopy-resolved structures, including 21 classified as either gain of function (GOF) or loss of function (LOF) in BK channel activity. Clusters of LOF variants were identified in the pore, the AC region (RCK1), and near the Ca2+ bowl (RCK2), overlapping with sites of pharmacological or endogenous modulation. However, no clustering was found for GOF variants. To further understand variants of uncertain significance (VUSs), assessments by multiple standard pathogenicity algorithms were compared, and new thresholds for sensitivity and specificity were established from confirmed GOF and LOF variants. An ensemble algorithm was constructed (KCNMA1 meta score (KMS)), consisting of a weighted summation of this trained dataset combined with a structural component derived from the Ca2+-bound and unbound BK channels. KMS assessment differed from the highest-performing individual algorithm (REVEL) at 10 VUS residues, and a subset were studied further by electrophysiology in HEK293 cells. M578T, E656A, and D965V (KMS+;REVEL-) were confirmed to alter BK channel properties in voltage-clamp recordings, and D800Y (KMS-;REVEL+) was assessed as benign under the test conditions. However, KMS failed to accurately assess K457E. These combined results reveal the distribution of potentially disease-causing KCNMA1 variants within BK channel functional domains and pathogenicity evaluation for VUSs, suggesting strategies for improving channel-level predictions in future studies by building on ensemble algorithms such as KMS.
Collapse
Affiliation(s)
- Hans J Moldenhauer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kelly Tammen
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
2
|
Van NTH, Kim WK, Nam JH. Challenges in the Therapeutic Targeting of KCa Channels: From Basic Physiology to Clinical Applications. Int J Mol Sci 2024; 25:2965. [PMID: 38474212 PMCID: PMC10932353 DOI: 10.3390/ijms25052965] [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: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024] Open
Abstract
Calcium-activated potassium (KCa) channels are ubiquitously expressed throughout the body and are able to regulate membrane potential and intracellular calcium concentrations, thereby playing key roles in cellular physiology and signal transmission. Consequently, it is unsurprising that KCa channels have been implicated in various diseases, making them potential targets for pharmaceutical interventions. Over the past two decades, numerous studies have been conducted to develop KCa channel-targeting drugs, including those for disorders of the central and peripheral nervous, cardiovascular, and urinary systems and for cancer. In this review, we synthesize recent findings regarding the structure and activating mechanisms of KCa channels. We also discuss the role of KCa channel modulators in therapeutic medicine. Finally, we identify the major reasons behind the delay in bringing these modulators to the pharmaceutical market and propose new strategies to promote their application.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| |
Collapse
|
3
|
Meredith AL. BK Channelopathies and KCNMA1-Linked Disease Models. Annu Rev Physiol 2024; 86:277-300. [PMID: 37906945 DOI: 10.1146/annurev-physiol-030323-042845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Novel KCNMA1 variants, encoding the BK K+ channel, are associated with a debilitating dyskinesia and epilepsy syndrome. Neurodevelopmental delay, cognitive disability, and brain and structural malformations are also diagnosed at lower incidence. More than half of affected individuals present with a rare negative episodic motor disorder, paroxysmal nonkinesigenic dyskinesia (PNKD3). The mechanistic relationship of PNKD3 to epilepsy and the broader spectrum of KCNMA1-associated symptomology is unknown. This review summarizes patient-associated KCNMA1 variants within the BK channel structure, functional classifications, genotype-phenotype associations, disease models, and treatment. Patient and transgenic animal data suggest delineation of gain-of-function (GOF) and loss-of-function KCNMA1 neurogenetic disease, validating two heterozygous alleles encoding GOF BK channels (D434G and N999S) as causing seizure and PNKD3. This discovery led to a variant-defined therapeutic approach for PNKD3, providing initial insight into the neurological basis. A comprehensive clinical definition of monogenic KCNMA1-linked disease and the neuronal mechanisms currently remain priorities for continued investigation.
Collapse
Affiliation(s)
- Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
4
|
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
|
5
|
Moldenhauer HJ, Tammen K, Meredith AL. Structural mapping of patient-associated KCNMA1 gene variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550850. [PMID: 37546746 PMCID: PMC10402178 DOI: 10.1101/2023.07.27.550850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
KCNMA1-linked channelopathy is a neurological disorder characterized by seizures, motor abnormalities, and neurodevelopmental disabilities. The disease mechanisms are predicted to result from alterations in KCNMA1-encoded BK K+ channel activity; however, only a subset of the patient-associated variants have been functionally studied. The localization of these variants within the tertiary structure or evaluation by pathogenicity algorithms has not been systematically assessed. In this study, 82 nonsynonymous patient-associated KCNMA1 variants were mapped within the BK channel protein. Fifty-three variants localized within cryo-EM resolved structures, including 21 classified as either gain-of-function (GOF) or loss-of-function (LOF) in BK channel activity. Clusters of LOF variants were identified in the pore, the AC region (RCK1), and near the Ca 2+ bowl (RCK2), overlapping with sites of pharmacological or endogenous modulation. However, no clustering was found for GOF variants. To further understand variants of uncertain significance (VUS), assessments by multiple standard pathogenicity algorithms were compared, and new thresholds for sensitivity and specificity were established from confirmed GOF and LOF variants. An ensemble algorithm was constructed (KCNMA1 Meta Score), consisting of a weighted summation of this trained dataset combined with a structural component derived from the Ca 2+ bound and unbound BK channels. KMS assessment differed from the highest performing individual algorithm (REVEL) at 10 VUS residues, and a subset were studied further by electrophysiology in HEK293 cells. M578T, E656A, and D965V (KMS+;REVEL-) were confirmed to alter BK channel properties in voltage-clamp recordings, and D800Y (KMS-;REVEL+) was assessed as benign under the test conditions. However, KMS failed to accurately assess K457E. These combined results reveal the distribution of potentially disease-causing KCNMA1 variants within BK channel functional domains and pathogenicity evaluation for VUS, suggesting strategies for improving channel-level predictions in future studies by building on ensemble algorithms such as KMS.
Collapse
|
6
|
Structure of the Human BK Ion Channel in Lipid Environment. MEMBRANES 2022; 12:membranes12080758. [PMID: 36005673 PMCID: PMC9414842 DOI: 10.3390/membranes12080758] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/27/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023]
Abstract
Voltage-gated and ligand-modulated ion channels play critical roles in excitable cells. To understand the interplay among voltage sensing, ligand binding, and channel opening, the structures of ion channels in various functional states and in lipid membrane environments need to be determined. Here, the random spherically constrained (RSC) single-particle cryo-EM method was employed to study human large conductance voltage- and calcium-activated potassium (hBK or hSlo1) channels reconstituted into liposomes. The hBK structure was determined at 3.5 Å resolution in the absence of Ca2+. Instead of the common fourfold symmetry observed in ligand-modulated ion channels, a twofold symmetry was observed in hBK in liposomes. Compared with the structure of isolated hSlo1 Ca2+ sensing gating rings, two opposing subunits in hBK unfurled, resulting in a wider opening towards the transmembrane region of hBK. In the pore gate domain, two opposing subunits also moved downwards relative to the two other subunits.
Collapse
|
7
|
Liang L, Liu H, Bartholdi D, van Haeringen A, Fernandez‐Jaén A, Peeters EEA, Xiong H, Bai X, Xu C, Ke T, Wang QK. Identification and functional analysis of two new de novo KCNMA1 variants associated with Liang-Wang syndrome. Acta Physiol (Oxf) 2022; 235:e13800. [PMID: 35156297 DOI: 10.1111/apha.13800] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023]
Abstract
AIM Loss-of-function KCNMA1 variants cause Liang-Wang syndrome (MIM #618729), a newly identified multiple malformation syndrome with a broad spectrum of developmental and neurological phenotypes. However, the full spectrum of clinical features and underlying pathogenic mechanisms need full elucidation. METHODS Exome sequencing was used to identify pathogenic variants. Patch-clamp recordings were performed to access the effects of KCNMA1 variants on BK channels. Total and membrane protein expression levels of BK channels were characterized using Western blotting. RESULTS We report identification and functional characterization of two new de novo loss-of-function KCNMA1 variants p.(A172T) and p.(A314T) with characteristics of Liang-Wang syndrome. Variant p.(A172T) is associated with developmental delay, cognitive impairment and ataxia. Mechanistically, p.(A172T) abolishes BK potassium current, inhibits Mg2+ -dependent gating, but shifts conductance-voltage (G-V) curves to more positive potentials when complexed with WT channels. Variant p.(A314T) is associated with developmental delay, intellectual disability, cognitive impairment, mild ataxia and generalized epilepsy; suppresses BK current amplitude; and shifts G-V curves to more positive potentials when expressed with WT channels. In addition, two new patients with previously reported gain-of-function variants p.(N536H) and p.(N995S) are found to show epilepsy and paroxysmal dyskinesia as reported previously, but also exhibit additional symptoms of cognitive impairment and dysmorphic features. Furthermore, variants p.(A314T) and p.(N536H) reduced total and membrane levels of BK proteins. CONCLUSION Our findings identified two new loss-of-function mutations of KCNMA1 associated with Liang-Wang syndrome, expanded the spectrum of clinical features associated with gain-of-function KCNMA1 variants and emphasized the overlapping features shared by gain-of-function and loss-of-function mutations.
Collapse
Affiliation(s)
- Lina Liang
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Huihui Liu
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Deborah Bartholdi
- Department of Human Genetics, Inselspital University Hospital Bern Bern Switzerland
| | - Arie van Haeringen
- Department of Clinical Genetics Leiden University Medical Center Leiden the Netherlands
| | - Alberto Fernandez‐Jaén
- Hospital Universitario Quirónsalud School of Medicine Universidad Europea de Madrid Madrid Spain
| | - Els E. A. Peeters
- Department of Child Neurology Juliana Children’s Hospital HAGA Medical Center The Hague the Netherlands
| | - Hongbo Xiong
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Xuemei Bai
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Chengqi Xu
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Tie Ke
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| | - Qing K. Wang
- Center for Human Genome Research Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology Huazhong University of Science and Technology Wuhan P. R. China
| |
Collapse
|
8
|
Ochoa SV, Otero L, Aristizabal-Pachon AF, Hinostroza F, Carvacho I, Torres YP. Hypoxic Regulation of the Large-Conductance, Calcium and Voltage-Activated Potassium Channel, BK. Front Physiol 2022; 12:780206. [PMID: 35002762 PMCID: PMC8727448 DOI: 10.3389/fphys.2021.780206] [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: 09/20/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022] Open
Abstract
Hypoxia is a condition characterized by a reduction of cellular oxygen levels derived from alterations in oxygen balance. Hypoxic events trigger changes in cell-signaling cascades, oxidative stress, activation of pro-inflammatory molecules, and growth factors, influencing the activity of various ion channel families and leading to diverse cardiovascular diseases such as myocardial infarction, ischemic stroke, and hypertension. The large-conductance, calcium and voltage-activated potassium channel (BK) has a central role in the mechanism of oxygen (O2) sensing and its activity has been related to the hypoxic response. BK channels are ubiquitously expressed, and they are composed by the pore-forming α subunit and the regulatory subunits β (β1–β4), γ (γ1–γ4), and LINGO1. The modification of biophysical properties of BK channels by β subunits underly a myriad of physiological function of these proteins. Hypoxia induces tissue-specific modifications of BK channel α and β subunits expression. Moreover, hypoxia modifies channel activation kinetics and voltage and/or calcium dependence. The reported effects on the BK channel properties are associated with events such as the increase of reactive oxygen species (ROS) production, increases of intracellular Calcium ([Ca2+]i), the regulation by Hypoxia-inducible factor 1α (HIF-1α), and the interaction with hemeproteins. Bronchial asthma, chronic obstructive pulmonary diseases (COPD), and obstructive sleep apnea (OSA), among others, can provoke hypoxia. Untreated OSA patients showed a decrease in BK-β1 subunit mRNA levels and high arterial tension. Treatment with continuous positive airway pressure (CPAP) upregulated β1 subunit mRNA level, decreased arterial pressures, and improved endothelial function coupled with a reduction in morbidity and mortality associated with OSA. These reports suggest that the BK channel has a role in the response involved in hypoxia-associated hypertension derived from OSA. Thus, this review aims to describe the mechanisms involved in the BK channel activation after a hypoxic stimulus and their relationship with disorders like OSA. A deep understanding of the molecular mechanism involved in hypoxic response may help in the therapeutic approaches to treat the pathological processes associated with diseases involving cellular hypoxia.
Collapse
Affiliation(s)
- Sara V Ochoa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Semillero de Investigación, Biofísica y Fisiología de Canales Iónicos, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Liliana Otero
- Center of Dental Research Dentistry Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Fernando Hinostroza
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación de Estudios Avanzados del Maule, CIEAM, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile.,Facultad de Ciencias de la Salud, Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Universidad Católica del Maule, Talca, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Semillero de Investigación, Biofísica y Fisiología de Canales Iónicos, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
9
|
North KC, Zhang M, Singh AK, Zaytseva D, Slayden AV, Bukiya AN, Dopico AM. Cholesterol inhibition of slo1 channels is Ca2+-dependent and can be mediated by either high-affinity Ca2+-sensing site in the slo1 cytosolic tail. Mol Pharmacol 2021; 101:132-143. [PMID: 34969832 DOI: 10.1124/molpharm.121.000392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022] Open
Abstract
Ca2+-/voltage-gated K+ channels of large conductance (BK) are expressed in the cell membranes of all excitable tissues. Currents mediated by BK channel-forming slo1 homotetramers are consistently inhibited by increases in membrane cholesterol (CLR). The molecular mechanisms leading to this CLR action, however, remain unknown. Slo1 channels are activated by increases in Ca2+ nearby Ca2+-recognition sites in the slo1 cytosolic tail: one high-affinity and one low-affinity sites locate to the Regulator of Conductance for K+ (RCK) 1 domain, while another high-affinity site locates within the RCK2 domain. Here we first evaluated the cross-talking between Ca2+ and CLR on the function of slo1 (cbv1 isoform) channels reconstituted into planar lipid bilayers. CLR robustly reduced channel open probability while barely decreasing unitary current amplitude, with CLR maximal effects being observed at 10-30 µM internal Ca2+ CLR actions were not only modulated by internal Ca2+ levels but also disappeared in absence of this divalent. Moreover, in absence of Ca2+, BK channel-activating concentrations of Mg2+ (10 mM) did not support CLR action. Next, we evaluated CLR actions on channels where the different Ca2+-sensing sites present in the slo1 cytosolic domain became nonfunctional via mutagenesis. CLR still reduced the activity of low-affinity Ca2+ (RCK1:E379A, E404A) mutants. In contrast, CLR became inefficacious when both high-affinity Ca2+ sites were mutated (RCK1:D367A,D372A, and RCK2:D899N,D900N,D901N,D902N,D903N), yet still was able to decrease the activity of each high-affinity site mutant. Therefore, BK channel inhibition by CLR selectively requires optimal levels of Ca2+ being recognized by either of the slo1 high-affinity Ca2+-sensing sites. Significance Statement Results reveal that the widely reported inhibition of BK (slo1) channels by membrane cholesterol requires a physiologically range of internal Ca2+ and is selectively linked to the two high-affinity Ca2+-sensing sites located in the cytosolic tail domain of slo1 proteins, which underscores that Ca2+ and cholesterol actions are allosterically coupled to the channel gate. Cholesterol modification of BK channel activity likely contributes to disruption of normal physiology by common health conditions that are triggered by disruption of cholesterol homeostasis.
Collapse
Affiliation(s)
| | - Man Zhang
- Shanghai Center for System Biomedicine, Shanghai Jiao Tong University, China
| | | | | | | | - Anna N Bukiya
- Pharmacology, The University of Tennessee Health Science Center, United States
| | - Alex M Dopico
- Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, United States
| |
Collapse
|
10
|
Ji W, Shi D, Shi S, Yang X, Chen Y, An H, Pang C. TMEM16A protein: calcium binding site and its activation mechanism. Protein Pept Lett 2021; 28:1338-1348. [PMID: 34749600 DOI: 10.2174/0929866528666211105112131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/08/2021] [Accepted: 09/18/2021] [Indexed: 11/22/2022]
Abstract
TMEM16A mediates calcium-activated transmembrane flow of chloride ion and a variety of physiological functions. The binding of cytoplasmic calcium ions of TMEM16A and the consequent conformational changes of it are the key issues to explore the relationship between its structure and function. In recent years, researchers have explored this issue through electrophysiological experiment, structure resolving, molecular dynamic simulation and other methods. The structures of TMEM16 family members resolved by cryo-Electron microscopy (cryo-EM) and X-ray crystallization provide the primarily basis for the investigation of the molecular mechanism of TMEM16A. However, the binding and activation mechanism of calcium ions in TMEM16A are still unclear and controversial. This review discusses four Ca2+ sensing sites of TMEM16A and analyze activation properties of TMEM16A by them, which will help to understand the structure-function relationship of TMEM16A and throw light on the molecular design targeting TMEM16A channel.
Collapse
Affiliation(s)
- Wanying Ji
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Donghong Shi
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Sai Shi
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Xiao Yang
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Yafei Chen
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Hailong An
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| | - Chunli Pang
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401. China
| |
Collapse
|
11
|
Lu T, Lee HC. Coronary Large Conductance Ca 2+-Activated K + Channel Dysfunction in Diabetes Mellitus. Front Physiol 2021; 12:750618. [PMID: 34744789 PMCID: PMC8567020 DOI: 10.3389/fphys.2021.750618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 11/24/2022] Open
Abstract
Diabetes mellitus (DM) is an independent risk of macrovascular and microvascular complications, while cardiovascular diseases remain a leading cause of death in both men and women with diabetes. Large conductance Ca2+-activated K+ (BK) channels are abundantly expressed in arteries and are the key ionic determinant of vascular tone and organ perfusion. It is well established that the downregulation of vascular BK channel function with reduced BK channel protein expression and altered intrinsic BK channel biophysical properties is associated with diabetic vasculopathy. Recent efforts also showed that diabetes-associated changes in signaling pathways and transcriptional factors contribute to the downregulation of BK channel expression. This manuscript will review our current understandings on the molecular, physiological, and biophysical mechanisms that underlie coronary BK channelopathy in diabetes mellitus.
Collapse
Affiliation(s)
- Tong Lu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hon-Chi Lee
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
12
|
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: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [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
|
13
|
A Calcium Sensor Discovered in Bluetongue Virus Nonstructural Protein 2 Is Critical for Virus Replication. J Virol 2020; 94:JVI.01099-20. [PMID: 32759321 PMCID: PMC7527055 DOI: 10.1128/jvi.01099-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022] Open
Abstract
After entering the host cells, viruses use cellular host factors to ensure a successful virus replication process. For replication in infected cells, members of the Reoviridae family form inclusion body-like structures known as viral inclusion bodies (VIB) or viral factories. Bluetongue virus (BTV) forms VIBs in infected cells through nonstructural protein 2 (NS2), a phosphoprotein. An important regulatory factor critical for VIB formation is phosphorylation of NS2. In our study, we discovered a characteristic calcium-binding EF-hand-like motif in NS2 and found that the calcium binding preferentially affects phosphorylation level of the NS2 and has a role in regulating VIB assembly. Many viruses use specific viral proteins to bind calcium ions (Ca2+) for stability or to modify host cell pathways; however, to date, no Ca2+ binding protein has been reported in bluetongue virus (BTV), the causative agent of bluetongue disease in livestock. Here, using a comprehensive bioinformatics screening, we identified a putative EF-hand-like Ca2+ binding motif in the carboxyl terminal region of BTV nonstructural phosphoprotein 2 (NS2). Subsequently, using a recombinant NS2, we demonstrated that NS2 binds Ca2+ efficiently and that Ca2+ binding was perturbed when the Asp and Glu residues in the motif were substituted by alanine. Using circular dichroism analysis, we found that Ca2+ binding by NS2 triggered a helix-to-coil secondary structure transition. Further, cryo-electron microscopy in the presence of Ca2+ revealed that NS2 forms helical oligomers which, when aligned with the N-terminal domain crystal structure, suggest an N-terminal domain that wraps around the C-terminal domain in the oligomer. Further, an in vitro kinase assay demonstrated that Ca2+ enhanced the phosphorylation of NS2 significantly. Importantly, mutations introduced at the Ca2+ binding site in the viral genome by reverse genetics failed to allow recovery of viable virus, and the NS2 phosphorylation level and assembly of viral inclusion bodies (VIBs) were reduced. Together, our data suggest that NS2 is a dedicated Ca2+ binding protein and that calcium sensing acts as a trigger for VIB assembly, which in turn facilitates virus replication and assembly. IMPORTANCE After entering the host cells, viruses use cellular host factors to ensure a successful virus replication process. For replication in infected cells, members of the Reoviridae family form inclusion body-like structures known as viral inclusion bodies (VIB) or viral factories. Bluetongue virus (BTV) forms VIBs in infected cells through nonstructural protein 2 (NS2), a phosphoprotein. An important regulatory factor critical for VIB formation is phosphorylation of NS2. In our study, we discovered a characteristic calcium-binding EF-hand-like motif in NS2 and found that the calcium binding preferentially affects phosphorylation level of the NS2 and has a role in regulating VIB assembly.
Collapse
|
14
|
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
|
15
|
Lorenzo-Ceballos Y, Carrasquel-Ursulaez W, Castillo K, Alvarez O, Latorre R. Calcium-driven regulation of voltage-sensing domains in BK channels. eLife 2019; 8:44934. [PMID: 31509109 PMCID: PMC6763263 DOI: 10.7554/elife.44934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 09/10/2019] [Indexed: 12/17/2022] Open
Abstract
Allosteric interactions between the voltage-sensing domain (VSD), the Ca2+-binding sites, and the pore domain govern the mammalian Ca2+- and voltage-activated K+ (BK) channel opening. However, the functional relevance of the crosstalk between the Ca2+- and voltage-sensing mechanisms on BK channel gating is still debated. We examined the energetic interaction between Ca2+ binding and VSD activation by investigating the effects of internal Ca2+ on BK channel gating currents. Our results indicate that Ca2+ sensor occupancy has a strong impact on VSD activation through a coordinated interaction mechanism in which Ca2+ binding to a single α-subunit affects all VSDs equally. Moreover, the two distinct high-affinity Ca2+-binding sites contained in the C-terminus domains, RCK1 and RCK2, contribute equally to decrease the free energy necessary to activate the VSD. We conclude that voltage-dependent gating and pore opening in BK channels is modulated to a great extent by the interaction between Ca2+ sensors and VSDs.
Collapse
Affiliation(s)
- Yenisleidy Lorenzo-Ceballos
- Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
16
|
Guo R, Huang X, Jin X, Yang J. [Diltiazem inhibits proliferation and motility of hepatocellular cells in vitro by downregulating calcium-activated chloride channel TMEM16A]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:818-823. [PMID: 33168514 DOI: 10.3969/j.issn.1673-4254.2018.07.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To assess the inhibitory effect of diltiazem, a calcium channel inhibitor, on the proliferation and mobility of human hepatocellular carcinoma cells in vitro and explore the possible mechanism. METHODS Two human hepatocellular carcinoma cell lines, MHCC97H and 7402, were treated with different concentrations (0-400 μmol/L) of diltiazem for 12, 24, or 48 h, and the changes in the cell proliferation and mobility were observed with MTT assay and wound healing assay, respectively. The changes in the expressions of calcium-activated chloride channel TMEM16A at mRNA and protein levels in the treated cells were detected using RT-PCR and immunocytochemistry. RESULTS Treatment with diltiazem obviously inhibited the proliferation and suppressed the mobility of MHCC97H and 7402 cells in a time- and concentration-dependent manner (P < 0.05). Treatment with 100 μmol/L diltiazem for 24 h significantly inhibited the proliferation of MHCC97H cells and down-regulated the mRNA and protein levels of TMEM16A. In 7402 cells, diltiazem treatment at 50 μmol/L for 48 h resulted in the most significant inhibitory effect on the cell proliferation and TMEM16A expressions. CONCLUSIONS Diltiazem can transiently inhibit the invasion of hepatocellular carcinoma cells in vitro possibly by down-regulating the expression of TMEM16A at both the mRNA and protein levels.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaozhong Huang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xueyuan Jin
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jun Yang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
17
|
Molecular determinants of Ca 2+ sensitivity at the intersubunit interface of the BK channel gating ring. Sci Rep 2018; 8:509. [PMID: 29323236 PMCID: PMC5765161 DOI: 10.1038/s41598-017-19029-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022] Open
Abstract
The large-conductance calcium-activated K+ (BK) channel contains two intracellular tandem Ca2+-sensing RCK domains (RCK1 and RCK2), which tetramerize into a Ca2+ gating ring that regulates channel opening by conformational expansion in response to Ca2+ binding. Interestingly, the gating ring’s intersubunit assembly interface harbors the RCK2 Ca2+-binding site, known as the Ca2+ bowl. The gating ring’s assembly interface is made in part by intersubunit coordination of a Ca2+ ion between the Ca2+ bowl and an RCK1 Asn residue, N449, and by apparent intersubunit electrostatic interactions between E955 in RCK2 and R786 and R790 in the RCK2 of the adjacent subunit. To understand the role of the intersubunit assembly interface in Ca2+ gating, we performed mutational analyses of these putative interacting residues in human BK channels. We found that N449, despite its role in Ca2+ coordination, does not set the channel’s Ca2+ sensitivity, whereas E955 is a determinant of Ca2+ sensitivity, likely through intersubunit electrostatic interactions. Our findings provide evidence that the intersubunit assembly interface contains molecular determinants of Ca2+-sensitivity in BK channels.
Collapse
|
18
|
Zhou Y, Yang H, Cui J, Lingle CJ. Threading the biophysics of mammalian Slo1 channels onto structures of an invertebrate Slo1 channel. J Gen Physiol 2017; 149:985-1007. [PMID: 29025867 PMCID: PMC5677106 DOI: 10.1085/jgp.201711845] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/20/2017] [Indexed: 11/24/2022] Open
Abstract
Zhou et al. consider the biophysics of large-conductance Ca2+-activated Slo1 channels in the context of Aplysia Slo1 structures. For those interested in the machinery of ion channel gating, the Ca2+ and voltage-activated BK K+ channel provides a compelling topic for investigation, by virtue of its dual allosteric regulation by both voltage and intracellular Ca2+ and because its large-single channel conductance facilitates detailed kinetic analysis. Over the years, biophysical analyses have illuminated details of the allosteric regulation of BK channels and revealed insights into the mechanism of BK gating, e.g., inner cavity size and accessibility and voltage sensor-pore coupling. Now the publication of two structures of an Aplysia californica BK channel—one liganded and one metal free—promises to reinvigorate functional studies and interpretation of biophysical results. The new structures confirm some of the previous functional inferences but also suggest new perspectives regarding cooperativity between Ca2+-binding sites and the relationship between voltage- and Ca2+-dependent gating. Here we consider the extent to which the two structures explain previous functional data on pore-domain properties, voltage-sensor motions, and divalent cation binding and activation of the channel.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Anesthesiology, Washington University School of Medicine, St. Louis MO
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University, St. Louis, MO
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis MO
| |
Collapse
|
19
|
Functional validation of Ca 2+-binding residues from the crystal structure of the BK ion channel. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:943-952. [PMID: 28966112 DOI: 10.1016/j.bbamem.2017.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/21/2017] [Accepted: 09/24/2017] [Indexed: 12/31/2022]
Abstract
BK channels are dually regulated by voltage and Ca2+, providing a cellular mechanism to couple electrical and chemical signalling. Intracellular Ca2+ concentration is sensed by a large cytoplasmic region in the channel known as "gating ring", which is formed by four tandems of regulator of conductance for K+ (RCK1 and RCK2) domains. The recent crystal structure of the full-length BK channel from Aplysia californica has provided new information about the residues involved in Ca2+ coordination at the high-affinity binding sites located in the RCK1 and RCK2 domains, as well as their cooperativity. Some of these residues have not been previously studied in the human BK channel. In this work we have investigated, through site directed mutagenesis and electrophysiology, the effects of these residues on channel activation by voltage and Ca2+. Our results demonstrate that the side chains of two non-conserved residues proposed to coordinate Ca2+ in the A. californica structure (G523 and E591) have no apparent functional role in the human BK Ca2+ sensing mechanism. Consistent with the crystal structure, our data indicate that in the human channel the conserved residue R514 participates in Ca2+ coordination in the RCK1 binding site. Additionally, this study provides functional evidence indicating that R514 also interacts with residues E902 and Y904 connected to the Ca2+ binding site in RCK2. Interestingly, it has been proposed that this interaction may constitute a structural correlate underlying the cooperative interactions between the two high-affinity Ca2+ binding sites regulating the Ca2+ dependent gating of the BK channel. This article is part of a Special Issue entitled: Beyond the Structure-Function Horizon of Membrane Proteins edited by Ute Hellmich, Rupak Doshi and Benjamin McIlwain.
Collapse
|
20
|
Wang W, Zhang X, Gao Q, Lawas M, Yu L, Cheng X, Gu M, Sahoo N, Li X, Li P, Ireland S, Meredith A, Xu H. A voltage-dependent K + channel in the lysosome is required for refilling lysosomal Ca 2+ stores. J Cell Biol 2017; 216:1715-1730. [PMID: 28468834 PMCID: PMC5461029 DOI: 10.1083/jcb.201612123] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/05/2017] [Accepted: 04/10/2017] [Indexed: 01/04/2023] Open
Abstract
Ion-dependent channels and transporters have been identified in lysosomes, including the V-ATPase H+ pump and transient receptor potential mucolipin channels (TRPMLs), the principle Ca2+ release channels in the lysosome, but much less is understood about the roles of Na+ and K+ in lysosomal physiology. Wang et al. describe a voltage-sensitive, Ca2+-activated K+ current in the lysosome (LysoKVCa) and show that LysoKVCa regulates lysosomal membrane potential and refilling of lysosomal Ca2+ stores. The resting membrane potential (Δψ) of the cell is negative on the cytosolic side and determined primarily by the plasma membrane’s selective permeability to K+. We show that lysosomal Δψ is set by lysosomal membrane permeabilities to Na+ and H+, but not K+, and is positive on the cytosolic side. An increase in juxta-lysosomal Ca2+ rapidly reversed lysosomal Δψ by activating a large voltage-dependent and K+-selective conductance (LysoKVCa). LysoKVCa is encoded molecularly by SLO1 proteins known for forming plasma membrane BK channels. Opening of single LysoKVCa channels is sufficient to cause the rapid, striking changes in lysosomal Δψ. Lysosomal Ca2+ stores may be refilled from endoplasmic reticulum (ER) Ca2+ via ER–lysosome membrane contact sites. We propose that LysoKVCa serves as the perilysosomal Ca2+ effector to prime lysosomes for the refilling process. Consistently, genetic ablation or pharmacological inhibition of LysoKVCa, or abolition of its Ca2+ sensitivity, blocks refilling and maintenance of lysosomal Ca2+ stores, resulting in lysosomal cholesterol accumulation and a lysosome storage phenotype.
Collapse
Affiliation(s)
- Wuyang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Qiong Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Maria Lawas
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Lu Yu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xiping Cheng
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Mingxue Gu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Nirakar Sahoo
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xinran Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ping Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109.,Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Stephen Ireland
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Andrea Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Giraldez T, Rothberg BS. Understanding the conformational motions of RCK gating rings. J Gen Physiol 2017; 149:431-441. [PMID: 28246116 PMCID: PMC5379921 DOI: 10.1085/jgp.201611726] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/17/2017] [Indexed: 12/16/2022] Open
Abstract
A timely review of the structural basis of Ca2+-activated K+ channel modulation by regulator of conduction of K+ (RCK) domains Regulator of conduction of K+ (RCK) domains are ubiquitous regulators of channel and transporter activity in prokaryotes and eukaryotes. In humans, RCK domains form an integral component of large-conductance calcium-activated K channels (BK channels), key modulators of nerve, muscle, and endocrine cell function. In this review, we explore how the study of RCK domains in bacterial and human channels has contributed to our understanding of the structural basis of channel function. This knowledge will be critical in identifying mechanisms that underlie BK channelopathies that lead to epilepsy and other diseases, as well as regions of the channel that might be successfully targeted to treat such diseases.
Collapse
Affiliation(s)
- Teresa Giraldez
- Department of Basic Medical Sciences, Institute of Biomedical Technologies and Centre for Biomedical Research of the Canary Islands, Universidad de La Laguna, La Laguna 38071, Spain
| | - Brad S Rothberg
- Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140
| |
Collapse
|
22
|
Latorre R, Castillo K, Carrasquel-Ursulaez W, Sepulveda RV, Gonzalez-Nilo F, Gonzalez C, Alvarez O. Molecular Determinants of BK Channel Functional Diversity and Functioning. Physiol Rev 2017; 97:39-87. [DOI: 10.1152/physrev.00001.2016] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Large-conductance Ca2+- and voltage-activated K+ (BK) channels play many physiological roles ranging from the maintenance of smooth muscle tone to hearing and neurosecretion. BK channels are tetramers in which the pore-forming α subunit is coded by a single gene ( Slowpoke, KCNMA1). In this review, we first highlight the physiological importance of this ubiquitous channel, emphasizing the role that BK channels play in different channelopathies. We next discuss the modular nature of BK channel-forming protein, in which the different modules (the voltage sensor and the Ca2+ binding sites) communicate with the pore gates allosterically. In this regard, we review in detail the allosteric models proposed to explain channel activation and how the models are related to channel structure. Considering their extremely large conductance and unique selectivity to K+, we also offer an account of how these two apparently paradoxical characteristics can be understood consistently in unison, and what we have learned about the conduction system and the activation gates using ions, blockers, and toxins. Attention is paid here to the molecular nature of the voltage sensor and the Ca2+ binding sites that are located in a gating ring of known crystal structure and constituted by four COOH termini. Despite the fact that BK channels are coded by a single gene, diversity is obtained by means of alternative splicing and modulatory β and γ subunits. We finish this review by describing how the association of the α subunit with β or with γ subunits can change the BK channel phenotype and pharmacology.
Collapse
Affiliation(s)
- Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Romina V. Sepulveda
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Fernando Gonzalez-Nilo
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Carlos Gonzalez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso and Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Universidad Andres Bello, Facultad de Ciencias Biologicas, Center for Bioinformatics and Integrative Biology, Avenida Republica 239, Santiago, Chile and Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
23
|
Kaczmarek LK, Aldrich RW, Chandy KG, Grissmer S, Wei AD, Wulff H. International Union of Basic and Clinical Pharmacology. C. Nomenclature and Properties of Calcium-Activated and Sodium-Activated Potassium Channels. Pharmacol Rev 2017; 69:1-11. [PMID: 28267675 PMCID: PMC11060434 DOI: 10.1124/pr.116.012864] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
A subset of potassium channels is regulated primarily by changes in the cytoplasmic concentration of ions, including calcium, sodium, chloride, and protons. The eight members of this subfamily were originally all designated as calcium-activated channels. More recent studies have clarified the gating mechanisms for these channels and have documented that not all members are sensitive to calcium. This article describes the molecular relationships between these channels and provides an introduction to their functional properties. It also introduces a new nomenclature that differentiates between calcium- and sodium-activated potassium channels.
Collapse
Affiliation(s)
- Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Richard W Aldrich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - K George Chandy
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Stephan Grissmer
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Aguan D Wei
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Heike Wulff
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| |
Collapse
|
24
|
Cryo-EM structure of the open high-conductance Ca 2+-activated K + channel. Nature 2016; 541:46-51. [PMID: 27974795 DOI: 10.1038/nature20608] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022]
Abstract
The Ca2+-activated K+ channel, Slo1, has an unusually large conductance and contains a voltage sensor and multiple chemical sensors. Dual activation by membrane voltage and Ca2+ renders Slo1 central to processes that couple electrical signalling to Ca2+-mediated events such as muscle contraction and neuronal excitability. Here we present the cryo-electron microscopy structure of a full-length Slo1 channel from Aplysia californica in the presence of Ca2+ and Mg2+ at a resolution of 3.5 Å. The channel adopts an open conformation. Its voltage-sensor domain adopts a non-domain-swapped attachment to the pore and contacts the cytoplasmic Ca2+-binding domain from a neighbouring subunit. Unique structural features of the Slo1 voltage sensor suggest that it undergoes different conformational changes than other known voltage sensors. The structure reveals the molecular details of three distinct divalent cation-binding sites identified through electrophysiological studies of mutant Slo1 channels.
Collapse
|
25
|
Diaz-Franulic I, Poblete H, Miño-Galaz G, González C, Latorre R. Allosterism and Structure in Thermally Activated Transient Receptor Potential Channels. Annu Rev Biophys 2016; 45:371-98. [DOI: 10.1146/annurev-biophys-062215-011034] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ignacio Diaz-Franulic
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago 8370146, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile; ,
- Fraunhofer Chile Research, Las Condes 7550296, Santiago, Chile
| | - Horacio Poblete
- Institute of Computational Comparative Medicine, Nanotechnology Innovation Center of Kansas State, Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas 66506-5802
| | - Germán Miño-Galaz
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago 8370146, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile; ,
| | - Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile; ,
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile; ,
| |
Collapse
|
26
|
Hoshi T, Heinemann SH. Modulation of BK Channels by Small Endogenous Molecules and Pharmaceutical Channel Openers. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:193-237. [PMID: 27238265 DOI: 10.1016/bs.irn.2016.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Voltage- and Ca(2+)-activated K(+) channels of big conductance (BK channels) are abundantly found in various organs and their relevance for smooth muscle tone and neuronal signaling is well documented. Dysfunction of BK channels is implicated in an array of human diseases involving many organs including the nervous, pulmonary, cardiovascular, renal, and urinary systems. In humans a single gene (KCNMA1) encodes the pore-forming α subunit (Slo1) of BK channels, but the channel properties are variable because of alternative splicing, tissue- and subcellular-specific auxiliary subunits (β, γ), posttranslational modifications, and a multitude of endogenous signaling molecules directly affecting the channel function. Initiatives to develop drugs capable of activating BK channels (channel openers) therefore need to consider the tissue-specific variability of BK channel structure and the potential interference with endogenously produced regulatory factors. The atomic structural basis of BK channel function is only beginning to be revealed. However, building on detailed knowledge of BK channel function, including its single-channel characteristics, voltage- and Ca(2+) dependence of channel gating, and modulation by diffusible messengers, a multi-tier allosteric model of BK channel gating (Horrigan and Aldrich (HA) model) has become a valuable tool in studying modulation of the channel. Using the conceptual framework of the HA model, we here review the functional impact of endogenous modulatory factors and select small synthetic compounds that regulate BK channel activity. Furthermore, we devise experimental approaches for studying BK channel-drug interactions with the aim to classify BK-modulating substances according to their molecular mode of action.
Collapse
Affiliation(s)
- T Hoshi
- University of Pennsylvania, Philadelphia, PA, United States.
| | - S H Heinemann
- Friedrich Schiller University Jena & Jena University Hospital, Jena, Germany
| |
Collapse
|
27
|
Abstract
BK channels are universal regulators of cell excitability, given their exceptional unitary conductance selective for K(+), joint activation mechanism by membrane depolarization and intracellular [Ca(2+)] elevation, and broad expression pattern. In this chapter, we discuss the structural basis and operational principles of their activation, or gating, by membrane potential and calcium. We also discuss how the two activation mechanisms interact to culminate in channel opening. As members of the voltage-gated potassium channel superfamily, BK channels are discussed in the context of archetypal family members, in terms of similarities that help us understand their function, but also seminal structural and biophysical differences that confer unique functional properties.
Collapse
Affiliation(s)
- A Pantazis
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - R Olcese
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
28
|
Humphries ESA, Dart C. Neuronal and Cardiovascular Potassium Channels as Therapeutic Drug Targets: Promise and Pitfalls. JOURNAL OF BIOMOLECULAR SCREENING 2015; 20:1055-73. [PMID: 26303307 PMCID: PMC4576507 DOI: 10.1177/1087057115601677] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/26/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Potassium (K(+)) channels, with their diversity, often tissue-defined distribution, and critical role in controlling cellular excitability, have long held promise of being important drug targets for the treatment of dysrhythmias in the heart and abnormal neuronal activity within the brain. With the exception of drugs that target one particular class, ATP-sensitive K(+) (KATP) channels, very few selective K(+) channel activators or inhibitors are currently licensed for clinical use in cardiovascular and neurological disease. Here we review what a range of human genetic disorders have told us about the role of specific K(+) channel subunits, explore the potential of activators and inhibitors of specific channel populations as a therapeutic strategy, and discuss possible reasons for the difficulty in designing clinically relevant K(+) channel modulators.
Collapse
Affiliation(s)
| | - Caroline Dart
- Institute of Integrative Biology, University of Liverpool, UK
| |
Collapse
|
29
|
Abstract
Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
Collapse
Affiliation(s)
- Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| | - Richard S Lewis
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
30
|
Takacs Z, Imredy JP, Bingham JP, Zhorov BS, Moczydlowski EG. Interaction of the BKCa channel gating ring with dendrotoxins. Channels (Austin) 2015; 8:421-32. [PMID: 25483585 DOI: 10.4161/19336950.2014.949186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Two classes of small homologous basic proteins, mamba snake dendrotoxins (DTX) and bovine pancreatic trypsin inhibitor (BPTI), block the large conductance Ca(2+)-activated K(+) channel (BKCa, KCa1.1) by production of discrete subconductance events when added to the intracellular side of the membrane. This toxin-channel interaction is unlikely to be pharmacologically relevant to the action of mamba venom, but as a fortuitous ligand-protein interaction, it has certain biophysical implications for the mechanism of BKCa channel gating. In this work we examined the subconductance behavior of 9 natural dendrotoxin homologs and 6 charge neutralization mutants of δ-dendrotoxin in the context of current structural information on the intracellular gating ring domain of the BKCa channel. Calculation of an electrostatic surface map of the BKCa gating ring based on the Poisson-Boltzmann equation reveals a predominantly electronegative surface due to an abundance of solvent-accessible side chains of negatively charged amino acids. Available structure-activity information suggests that cationic DTX/BPTI molecules bind by electrostatic attraction to site(s) on the gating ring located in or near the cytoplasmic side portals where the inactivation ball peptide of the β2 subunit enters to block the channel. Such an interaction may decrease the apparent unitary conductance by altering the dynamic balance of open versus closed states of BKCa channel activation gating.
Collapse
|
31
|
Yang H, Zhang G, Cui J. BK channels: multiple sensors, one activation gate. Front Physiol 2015; 6:29. [PMID: 25705194 PMCID: PMC4319557 DOI: 10.3389/fphys.2015.00029] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/19/2015] [Indexed: 01/01/2023] Open
Abstract
Ion transport across cell membranes is essential to cell communication and signaling. Passive ion transport is mediated by ion channels, membrane proteins that create ion conducting pores across cell membrane to allow ion flux down electrochemical gradient. Under physiological conditions, majority of ion channel pores are not constitutively open. Instead, structural region(s) within these pores breaks the continuity of the aqueous ion pathway, thereby serves as activation gate(s) to control ions flow in and out. To achieve spatially and temporally regulated ion flux in cells, many ion channels have evolved sensors to detect various environmental stimuli or the metabolic states of the cell and trigger global conformational changes, thereby dynamically operate the opening and closing of their activation gate. The sensors of ion channels can be broadly categorized as chemical sensors and physical sensors to respond to chemical (such as neural transmitters, nucleotides and ions) and physical (such as voltage, mechanical force and temperature) signals, respectively. With the rapidly growing structural and functional information of different types of ion channels, it is now critical to understand how ion channel sensors dynamically control their gates at molecular and atomic level. The voltage and Ca2+ activated BK channels, a K+ channel with an electrical sensor and multiple chemical sensors, provide a unique model system for us to understand how physical and chemical energy synergistically operate its activation gate.
Collapse
Affiliation(s)
- Huanghe Yang
- Ion Channel Research Unit, Duke University Medical Center Durham, NC, USA ; Department of Biochemistry, Duke University Medical Center Durham, NC, USA
| | - Guohui Zhang
- Department of Biomedical Engineering, Washington University in Saint Louis St. Louis, MO, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in Saint Louis St. Louis, MO, USA ; Cardiac Bioelectricity and Arrhythmia Center, Washington University in Saint Louis St. Louis, MO, USA ; Center for The Investigation of Membrane Excitability Disorders, Washington University in Saint Louis St. Louis, MO, USA
| |
Collapse
|
32
|
Molecular mechanisms underlying the effect of the novel BK channel opener GoSlo: involvement of the S4/S5 linker and the S6 segment. Proc Natl Acad Sci U S A 2015; 112:2064-9. [PMID: 25653338 DOI: 10.1073/pnas.1400555112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
GoSlo-SR-5-6 is a novel large-conductance Ca(2+)-activated K(+) (BK) channel agonist that shifts the activation V1/2 of these channels in excess of -100 mV when applied at a concentration of 10 μM. Although the structure-activity relationship of this family of molecules has been established, little is known about how they open BK channels. To help address this, we used a combination of electrophysiology, mutagenesis, and mathematical modeling to investigate the molecular mechanisms underlying the effect of GoSlo-SR-5-6. Our data demonstrate that the effects of this agonist are practically abolished when three point mutations are made: L227A in the S4/S5 linker in combination with S317R and I326A in the S6C region. Our data suggest that GoSlo-SR-5-6 interacts with the transmembrane domain of the channel to enhance pore opening. The Horrigan-Aldrich model suggests that GoSlo-SR-5-6 works by stabilizing the open conformation of the channel and the activated state of the voltage sensors, yet decouples the voltage sensors from the pore gate.
Collapse
|
33
|
A charged residue in S4 regulates coupling among the activation gate, voltage, and Ca2+ sensors in BK channels. J Neurosci 2015; 34:12280-8. [PMID: 25209270 DOI: 10.1523/jneurosci.1174-14.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Coupling between the activation gate and sensors of physiological stimuli during ion channel activation is an important, but not well-understood, molecular process. One difficulty in studying sensor-gate coupling is to distinguish whether a structural perturbation alters the function of the sensor, the gate, or their coupling. BK channels are activated by membrane voltage and intracellular Ca(2+) via allosteric mechanisms with coupling among the activation gate and sensors quantitatively defined, providing an excellent model system for studying sensor-gate coupling. By studying BK channels expressed in Xenopus oocytes, here we show that mutation E219R in S4 alters channel function by two independent mechanisms: one is to change voltage sensor activation, shifting voltage dependence, and increase valence of gating charge movements; the other is to regulate coupling among the activation gate, voltage sensor, and Ca(2+) binding via electrostatic interactions with E321/E324 located in the cytosolic side of S6 in a neighboring subunit, resulting in a shift of the voltage dependence of channel opening and increased Ca(2+) sensitivity. These results suggest a structural arrangement of the inner pore of BK channels differing from that in other voltage gated channels.
Collapse
|
34
|
Tang QY, Zhang Z, Meng XY, Cui M, Logothetis DE. Structural determinants of phosphatidylinositol 4,5-bisphosphate (PIP2) regulation of BK channel activity through the RCK1 Ca2+ coordination site. J Biol Chem 2014; 289:18860-72. [PMID: 24778177 DOI: 10.1074/jbc.m113.538033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Big or high conductance potassium (BK) channels are activated by voltage and intracellular calcium (Ca(2+)). Phosphatidylinositol 4,5-bisphosphate (PIP2), a ubiquitous modulator of ion channel activity, has been reported to enhance Ca(2+)-driven gating of BK channels, but a molecular understanding of this interplay or even of the PIP2 regulation of this channel's activity remains elusive. Here, we identify structural determinants in the KDRDD loop (which follows the αA helix in the RCK1 domain) to be responsible for the coupling between Ca(2+) and PIP2 in regulating BK channel activity. In the absence of Ca(2+), RCK1 structural elements limit channel activation through a decrease in the channel's PIP2 apparent affinity. This inhibitory influence of BK channel activation can be relieved by mutation of residues that (a) connect either the RCK1 Ca(2+) coordination site (Asp(367) or its flanking basic residues in the KDRDD loop) to the PIP2-interacting residues (Lys(392) and Arg(393)) found in the αB helix or (b) are involved in hydrophobic interactions between the αA and αB helix of the RCK1 domain. In the presence of Ca(2+), the RCK1-inhibitory influence of channel-PIP2 interactions and channel activity is relieved by Ca(2+) engaging Asp(367). Our results demonstrate that, along with Ca(2+) and voltage, PIP2 is a third factor critical to the integral control of BK channel activity.
Collapse
Affiliation(s)
- Qiong-Yao Tang
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298 and the Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou 221004, China
| | - Zhe Zhang
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298 and the Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou 221004, China
| | - Xuan-Yu Meng
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298 and
| | - Meng Cui
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298 and
| | - Diomedes E Logothetis
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298 and
| |
Collapse
|
35
|
Abstract
TMEM16 proteins, also known as anoctamins, are involved in a variety of functions that include ion transport, phospholipid scrambling, and regulation of other membrane proteins. The first two members of the family, TMEM16A (anoctamin-1, ANO1) and TMEM16B (anoctamin-2, ANO2), function as Ca2+-activated Cl- channels (CaCCs), a type of ion channel that plays important functions such as transepithelial ion transport, smooth muscle contraction, olfaction, phototransduction, nociception, and control of neuronal excitability. Genetic ablation of TMEM16A in mice causes impairment of epithelial Cl- secretion, tracheal abnormalities, and block of gastrointestinal peristalsis. TMEM16A is directly regulated by cytosolic Ca2+ as well as indirectly by its interaction with calmodulin. Other members of the anoctamin family, such as TMEM16C, TMEM16D, TMEM16F, TMEM16G, and TMEM16J, may work as phospholipid scramblases and/or ion channels. In particular, TMEM16F (ANO6) is a major contributor to the process of phosphatidylserine translocation from the inner to the outer leaflet of the plasma membrane. Intriguingly, TMEM16F is also associated with the appearance of anion/cation channels activated by very high Ca2+ concentrations. Furthermore, a TMEM16 protein expressed in Aspergillus fumigatus displays both ion channel and lipid scramblase activity. This finding suggests that dual function is an ancestral characteristic of TMEM16 proteins and that some members, such as TMEM16A and TMEM16B, have evolved to a pure channel function. Mutations in anoctamin genes (ANO3, ANO5, ANO6, and ANO10) cause various genetic diseases. These diseases suggest the involvement of anoctamins in a variety of cell functions whose link with ion transport and/or lipid scrambling needs to be clarified.
Collapse
|
36
|
Brenker C, Zhou Y, Müller A, Echeverry FA, Trötschel C, Poetsch A, Xia XM, Bönigk W, Lingle CJ, Kaupp UB, Strünker T. The Ca2+-activated K+ current of human sperm is mediated by Slo3. eLife 2014; 3:e01438. [PMID: 24670955 PMCID: PMC3966514 DOI: 10.7554/elife.01438] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sperm are equipped with a unique set of ion channels that orchestrate fertilization. In mouse sperm, the principal K+ current (IKSper) is carried by the Slo3 channel, which sets the membrane potential (Vm) in a strongly pHi-dependent manner. Here, we show that IKSper in human sperm is activated weakly by pHi and more strongly by Ca2+. Correspondingly, Vm is strongly regulated by Ca2+ and less so by pHi. We find that inhibitors of Slo3 suppress human IKSper, and we identify the Slo3 protein in the flagellum of human sperm. Moreover, heterologously expressed human Slo3, but not mouse Slo3, is activated by Ca2+ rather than by alkaline pHi; current–voltage relations of human Slo3 and human IKSper are similar. We conclude that Slo3 represents the principal K+ channel in human sperm that carries the Ca2+-activated IKSper current. We propose that, in human sperm, the progesterone-evoked Ca2+ influx carried by voltage-gated CatSper channels is limited by Ca2+-controlled hyperpolarization via Slo3. DOI:http://dx.doi.org/10.7554/eLife.01438.001 A sperm that has been ejaculated into the female reproductive tract must complete a number of tasks to pass on its genes to the next generation. First it must travel along a meandering route to encounter an egg, before pushing through a jelly-like coating that surrounds the egg and then, finally, fusing with the egg’s surface membrane. In order to complete these steps and fertilise the egg, a sperm must undergo a process called ‘capacitation’. This process, and a variety of other sperm functions, involves the controlled flux of positive ions into and out of the sperm via specific ion channels that are located in the cell membrane. The properties of the ion channels that allow protons and calcium ions to move into and out of human sperm are well understood, but less is known about the channels that control the movement of potassium ions. In mice, a channel called Slo3 allows potassium ions to flow out of the sperm and makes the membrane voltage of these cells more negative. Also, in mice, this channel is essential for the sperm to function correctly, and for fertilization. However, in humans, it is unclear if the Slo3 channel is present in sperm and if it performs the same role. Now, Brenker et al. have shown that the flow of potassium ions out of human sperm occurs via the Slo3 channel, and that human Slo3 is responsible for setting the membrane voltage of these cells. However, whereas the mouse Slo3 channel is opened in response to a decrease in the concentration of protons within the sperm (i.e., an increase of the pH inside the cell), human Slo3 is largely controlled by changes in the levels of calcium ions. An increase in the calcium concentration within the cell opens the human Slo3 channel, more than a decrease in the proton concentration does. Altogether, Brenker et al. identify Slo3 as the principal potassium channel in human sperm and reveal more fundamental differences between human sperm and mouse sperm. Thereby, this work further stresses the need to be cautious about using mice as a model of male fertility in humans. DOI:http://dx.doi.org/10.7554/eLife.01438.002
Collapse
Affiliation(s)
- Christoph Brenker
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Ca(2+)-activated Cl(-) channels (CaCCs) are plasma membrane proteins involved in various important physiological processes. In epithelial cells, CaCC activity mediates the secretion of Cl(-) and of other anions, such as bicarbonate and thiocyanate. In smooth muscle and excitable cells of the nervous system, CaCCs have an excitatory role coupling intracellular Ca(2+) elevation to membrane depolarization. Recent studies indicate that TMEM16A (transmembrane protein 16 A or anoctamin 1) and TMEM16B (transmembrane protein 16 B or anoctamin 2) are CaCC-forming proteins. Induced expression of TMEM16A and B in null cells by transfection causes the appearance of Ca(2+)-activated Cl(-) currents similar to those described in native tissues. Furthermore, silencing of TMEM16A by RNAi causes disappearance of CaCC activity in cells from airway epithelium, biliary ducts, salivary glands, and blood vessel smooth muscle. Mice devoid of TMEM16A expression have impaired Ca(2+)-dependent Cl(-) secretion in the epithelial cells of the airways, intestine, and salivary glands. These animals also show a loss of gastrointestinal motility, a finding consistent with an important function of TMEM16A in the electrical activity of gut pacemaker cells, that is, the interstitial cells of Cajal. Identification of TMEM16 proteins will help to elucidate the molecular basis of Cl(-) transport.
Collapse
Affiliation(s)
- Loretta Ferrera
- Laboratory of Molecular Genetics, Istituto Giannina Gaslini, Genova, Italy
| | | | | |
Collapse
|
38
|
Tian C, Zhu R, Zhu L, Qiu T, Cao Z, Kang T. Potassium Channels: Structures, Diseases, and Modulators. Chem Biol Drug Des 2013; 83:1-26. [DOI: 10.1111/cbdd.12237] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Chuan Tian
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| | - Ruixin Zhu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Lixin Zhu
- Department of Pediatrics; Digestive Diseases and Nutrition Center; The State University of New York at Buffalo; Buffalo NY 14226 USA
| | - Tianyi Qiu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Zhiwei Cao
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Tingguo Kang
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| |
Collapse
|
39
|
Hoshi T, Pantazis A, Olcese R. Transduction of voltage and Ca2+ signals by Slo1 BK channels. Physiology (Bethesda) 2013; 28:172-89. [PMID: 23636263 DOI: 10.1152/physiol.00055.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Large-conductance Ca2+ -and voltage-gated K+ channels are activated by an increase in intracellular Ca2+ concentration and/or depolarization. The channel activation mechanism is well described by an allosteric model encompassing the gate, voltage sensors, and Ca2+ sensors, and the model is an excellent framework to understand the influences of auxiliary β and γ subunits and regulatory factors such as Mg2+. Recent advances permit elucidation of structural correlates of the biophysical mechanism.
Collapse
Affiliation(s)
- T Hoshi
- Department of Physiology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
40
|
Horrigan FT. Perspectives on: conformational coupling in ion channels: conformational coupling in BK potassium channels. ACTA ACUST UNITED AC 2013. [PMID: 23183698 PMCID: PMC3514727 DOI: 10.1085/jgp.201210849] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Large conductance calcium- and voltage-dependent BK potassium channels (aka BK(Ca), MaxiK, Slo1, KCa1.1, and KCNMA1) are expressed in a wide variety of tissues throughout the body and are activated by both intracellular Ca(2+) and membrane depolarization. Owing to these properties, BK channels participate in diverse physiological processes from electrical excitability in neurons and secretory cells, and regulation of smooth muscle tone to tuning of auditory hair cells (Vergara et al., 1998; Ghatta et al., 2006). The response to voltage and Ca(2+) allows BK channels to integrate electrical and calcium signaling, which is central to their physiological role. Understanding how BK and other multimodal channels are regulated by and integrate diverse stimuli is not only physiologically important but also relevant to the topic of conformational coupling. As a voltage- and ligand-dependent channel, BK channels contain both voltage-sensor and ligand-binding domains as well as a gate to regulate the flow of K(+) through the pore. Coupling of conformational changes in one domain to another provides the basis for transducing voltage and ligand binding into channel opening and, therefore, defines, together with the functional properties of the gate and sensors, the signal transduction properties of the channel. The goal of this perspective is to provide an overview on the role and molecular basis of conformational coupling between functional domains in BK channels and outline some of the questions that remain to be answered.
Collapse
Affiliation(s)
- Frank T Horrigan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX 77030, USA.
| |
Collapse
|
41
|
Contreras GF, Castillo K, Enrique N, Carrasquel-Ursulaez W, Castillo JP, Milesi V, Neely A, Alvarez O, Ferreira G, González C, Latorre R. A BK (Slo1) channel journey from molecule to physiology. Channels (Austin) 2013; 7:442-58. [PMID: 24025517 DOI: 10.4161/chan.26242] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium and voltage-activated potassium (BK) channels are key actors in cell physiology, both in neuronal and non-neuronal cells and tissues. Through negative feedback between intracellular Ca (2+) and membrane voltage, BK channels provide a damping mechanism for excitatory signals. Molecular modulation of these channels by alternative splicing, auxiliary subunits and post-translational modifications showed that these channels are subjected to many mechanisms that add diversity to the BK channel α subunit gene. This complexity of interactions modulates BK channel gating, modifying the energetic barrier of voltage sensor domain activation and channel opening. Regions for voltage as well as Ca (2+) sensitivity have been identified, and the crystal structure generated by the 2 RCK domains contained in the C-terminal of the channel has been described. The linkage of these channels to many intracellular metabolites and pathways, as well as their modulation by extracellular natural agents, has been found to be relevant in many physiological processes. This review includes the hallmarks of BK channel biophysics and its physiological impact on specific cells and tissues, highlighting its relationship with auxiliary subunit expression.
Collapse
Affiliation(s)
- Gustavo F Contreras
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Doctorado en Ciencias mención Neurociencia; Universidad de Valparaíso; Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | - Nicolás Enrique
- Grupo de Investigación en Fisiología Vascular (GINFIV); Universidad Nacional de la Plata; La Plata, Argentina
| | - Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Doctorado en Ciencias mención Neurociencia; Universidad de Valparaíso; Valparaíso, Chile
| | - Juan Pablo Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Facultad de Ciencias; Universidad de Chile; Santiago, Chile
| | - Verónica Milesi
- Grupo de Investigación en Fisiología Vascular (GINFIV); Universidad Nacional de la Plata; La Plata, Argentina
| | - Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | | | - Gonzalo Ferreira
- Laboratorio de Canales Iónicos; Departamento de Biofísica; Facultad de Medicina; Universidad de la República; Montevideo, Uruguay
| | - Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| |
Collapse
|
42
|
Pang C, Cao T, Li J, Jia M, Zhang S, Ren S, An H, Zhan Y. Combining fragment homology modeling with molecular dynamics aims at prediction of Ca²⁺ binding sites in CaBPs. J Comput Aided Mol Des 2013; 27:697-705. [PMID: 23934058 DOI: 10.1007/s10822-013-9668-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/31/2013] [Indexed: 11/28/2022]
Abstract
The family of calcium-binding proteins (CaBPs) consists of dozens of members and contributes to all aspects of the cell's function, from homeostasis to learning and memory. However, the Ca²⁺-binding mechanism is still unclear for most of CaBPs. To identify the Ca²⁺-binding sites of CaBPs, this study presented a computational approach which combined the fragment homology modeling with molecular dynamics simulation. For validation, we performed a two-step strategy as follows: first, the approach is used to identify the Ca²⁺-binding sites of CaBPs, which have the EF-hand Ca²⁺-binding site and the detailed binding mechanism. To accomplish this, eighteen crystal structures of CaBPs with 49 Ca²⁺-binding sites are selected to be analyzed including calmodulin. The computational method identified 43 from 49 Ca²⁺-binding sites. Second, we performed the approach to large-conductance Ca²⁺-activated K⁺ (BK) channels which don't have clear Ca²⁺-binding mechanism. The simulated results are consistent with the experimental data. The computational approach may shed some light on the identification of Ca²⁺-binding sites in CaBPs.
Collapse
Affiliation(s)
- ChunLi Pang
- Institute of Biophysics, Hebei University of Technology, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Molecular mechanisms of large-conductance ca (2+) -activated potassium channel activation by ginseng gintonin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:323709. [PMID: 23662129 PMCID: PMC3638619 DOI: 10.1155/2013/323709] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/05/2013] [Accepted: 03/12/2013] [Indexed: 01/12/2023]
Abstract
Gintonin is a unique lysophosphatidic acid (LPA) receptor ligand
found in Panax ginseng. Gintonin induces transient
[Ca2+]i
through G protein-coupled LPA receptors. Large-conductance Ca2+-activated
K+ (BKCa)
channels are expressed in blood vessels and neurons and
play important roles in blood vessel relaxation and attenuation of
neuronal excitability. BKCa channels are activated by transient
[Ca2+]i
and are regulated by various Ca2+-dependent kinases. We
investigated the molecular mechanisms of BKCa channel activation
by gintonin. BKCa channels are heterologously expressed in
Xenopus oocytes. Gintonin treatment induced BKCa channel activation in
oocytes expressing the BKCa channel α subunit in a
concentration-dependent manner (EC50 = 0.71 ± 0.08 µg/mL).
Gintonin-mediated BKCa channel activation was blocked by a PKC
inhibitor, calphostin, and by the calmodulin inhibitor,
calmidazolium. Site-directed mutations in BKCa channels targeting
CaM kinase II or PKC phosphorylation sites but not PKA
phosphorylation sites attenuated gintonin action. Mutations in the
Ca2+ bowl and the regulator of K+ conductance (RCK) site also
blocked gintonin action. These results indicate that
gintonin-mediated BKCa channel activations are achieved through
LPA1 receptor-phospholipase C-IP3-Ca2+-PKC-calmodulin-CaM kinase
II pathways and calcium binding to the Ca2+ bowl and RCK domain.
Gintonin could be a novel contributor against blood vessel
constriction and over-excitation of neurons.
Collapse
|
44
|
State-dependent FRET reports calcium- and voltage-dependent gating-ring motions in BK channels. Proc Natl Acad Sci U S A 2013; 110:5217-22. [PMID: 23479636 DOI: 10.1073/pnas.1219611110] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Large-conductance voltage- and calcium-dependent potassium channels (BK, "Big K+") are important controllers of cell excitability. In the BK channel, a large C-terminal intracellular region containing a "gating-ring" structure has been proposed to transduce Ca(2+) binding into channel opening. Using patch-clamp fluorometry, we have investigated the calcium and voltage dependence of conformational changes of the gating-ring region of BK channels, while simultaneously monitoring channel conductance. Fluorescence resonance energy transfer (FRET) between fluorescent protein inserts indicates that Ca(2+) binding produces structural changes of the gating ring that are much larger than those predicted by current X-ray crystal structures of isolated gating rings.
Collapse
|
45
|
Carvalho-de-Souza JL, Varanda WA, Tostes RC, Chignalia AZ. BK Channels in Cardiovascular Diseases and Aging. Aging Dis 2013; 4:38-49. [PMID: 23423545 PMCID: PMC3570140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/03/2012] [Accepted: 12/04/2012] [Indexed: 06/01/2023] Open
Abstract
Aging is a major risk factor for cardiovascular diseases, one of the main world-wide causes of death. Several structural and functional changes occur in the cardiovascular system during the aging process and the mechanisms involved in such alterations are yet to be completely described. BK channels are transmembrane proteins that play a key role in many physiological processes, including regulation of vascular tone. In vascular smooth muscle cells, BK opening and the consequent efflux of potassium (K(+)) leads to membrane hyperpolarization, which is followed by the closure of voltage-dependent Ca(2+) channels, reduction of Ca(2+) entry and vasodilatation. BK regulates nitric oxide-mediated vasodilatation and thus is crucial for normal endothelial function. Herein we will briefly review general structural properties of BK and focus on their function in the cardiovascular system emphasizing their role in cardiovascular aging and diseases.
Collapse
Affiliation(s)
| | | | - Rita C. Tostes
- Department of Pharmacology School of Medicine of Ribeirao Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Andreia Z. Chignalia
- Department of Pharmacology School of Medicine of Ribeirao Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
46
|
Levitsky DO, Takahashi M. Interplay of Ca(2+) and Mg (2+) in sodium-calcium exchanger and in other Ca(2+)-binding proteins: magnesium, watchdog that blocks each turn if able. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 961:65-78. [PMID: 23224871 DOI: 10.1007/978-1-4614-4756-6_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Sodium-calcium exchange across plasma membrane is regulated by intracellular calcium ions. The sodium-calcium exchanger (NCX1) is activated by successive saturation of numerous Ca(2+)-binding sites located in the intracellular loop of the protein. The progressive saturation of the binding domain CBD12 by Ca(2+) results in a series of conformational changes of CBD12 as well as of entire NCX1 molecule. Like other soluble and membrane Ca(2+)-binding proteins, NCX1 can also be regulated by Mg(2+) that antagonises Ca(2+) at the level of divalent cation-binding sites. This chapter summarises data on Mg(2+) impacts in the cells. Regulatory action of Mg(2+) on intracellular Ca(2+)-dependent processes can be achieved due to changes of its cytoplasmic level, which take place in the range of [Mg(2+)](i) from 0.5 to 3 mM. Under normal conditions, these changes are ensured by activation of plasmalemmal Mg(2+) transport systems and by variations in ATP level in cytoplasm. In heart and in brain, some pathological conditions, such as hypoxia, ischemia and ischemia followed by reperfusion, are associated with an important increase in intracellular Ca(2+). The tissue damage due to Ca(2+) overload may be prevented by Mg(2+). The protective actions of Mg(2+) can be achieved due to its ability to compete with Ca(2+) for the binding sites in a number of proteins responsible for the rise in intracellular free Ca(2+), including NCX1, in case when the reverse mode of Na(+)/Ca(2+) exchange becomes predominant. Saturation of CBD12 by Mg(2+) results in important changes of NCX1 conformation. Modulating actions of Mg(2+) on the conformation of NCX1 were detected at a narrow range of Mg(2+) concentration, from 0.5 to 1 mM. These data support an idea that variations of intracellular Mg(2+) could modify transmembrane Ca(2+) movements ensured by NCX1.
Collapse
Affiliation(s)
- Dmitri O Levitsky
- Unité de Fonctionnalité et Ingénierie des Protéines, Université de Nantes, Nantes Cedex 03, France.
| | | |
Collapse
|
47
|
Smith FJ, Rothberg BS. Analysis of Ca(2+)-binding sites in the MthK RCK domain by X-ray crystallography. Methods Mol Biol 2013; 998:277-287. [PMID: 23529438 DOI: 10.1007/978-1-62703-351-0_22] [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] [Indexed: 06/02/2023]
Abstract
Regulator of K(+) conductance (RCK) domains form a conserved class of ligand-binding domains that control the activity of a variety of prokaryotic and eukaryotic K(+) channels. Structural analysis of these domains by X-ray crystallography has provided insight toward mechanisms underlying ligand binding and channel gating, and thus the experimental strategies aimed at determining structures of liganded and unliganded forms of the domains may be useful in analysis of other ligand-binding domains. Here, we describe a basic strategy for crystallographic analysis of the RCK domain from the MthK channel, for determination of its Ca(2+)-bound structure.
Collapse
Affiliation(s)
- Frank J Smith
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
48
|
Kong C, Zeng W, Ye S, Chen L, Sauer DB, Lam Y, Derebe MG, Jiang Y. Distinct gating mechanisms revealed by the structures of a multi-ligand gated K(+) channel. eLife 2012; 1:e00184. [PMID: 23240087 PMCID: PMC3510474 DOI: 10.7554/elife.00184] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 10/19/2012] [Indexed: 11/29/2022] Open
Abstract
The gating ring-forming RCK domain regulates channel gating in response to various cellular chemical stimuli in eukaryotic Slo channel families and the majority of ligand-gated prokaryotic K+ channels and transporters. Here we present structural and functional studies of a dual RCK-containing, multi-ligand gated K+ channel from Geobacter sulfurreducens, named GsuK. We demonstrate that ADP and NAD+ activate the GsuK channel, whereas Ca2+ serves as an allosteric inhibitor. Multiple crystal structures elucidate the structural basis of multi-ligand gating in GsuK, and also reveal a unique ion conduction pore with segmented inner helices. Structural comparison leads us to propose a novel pore opening mechanics that is distinct from other K+ channels. DOI:http://dx.doi.org/10.7554/eLife.00184.001 Most cells are surrounded by a semipermeable membrane, and although this membrane allows very few molecules to pass through it, cells can use transmembrane proteins to overcome this barrier. Some of these proteins import glucose, amino acids and other nutrients into the cell, while others transport ions into or out of the cell. Ion transport across the cell membrane is essential for a wide variety of biological processes, including signal transduction and the generation of electrical impulses in nerve cells. The pores that allow ions to travel through the cell membrane are known as ion channels, and most channels allow only one type of ion—usually sodium, calcium or potassium (K+) ions—to pass through them. There are many different types of ion channels and they are classified according to the type of ion they allow to pass through them, and by the gating mechanism that is used to open and close the channel. For example, ligand-gated K+ channels facilitate the passage of potassium ions and are opened and closed by ligands binding and unbinding to and from the channel. Most K+ channels are made up of four identical subunits, and in the majority of ligand-gated K+ channels in prokaryotes, each of these subunits will have one or two ligand-binding RCK domains (where RCK stands for regulating the conductance of K+). This is also true for some K+ channels in eukaryotes. While it is known that RCK domains are responsible for regulating the transport of potassium ions across the cell membranes of diverse organisms, little is known about the structure or gating mechanisms of K+ channels that are gated by more than one ligand. Kong et al. have studied a ligand-gated K+ channel called GsuK that has two RCK domains per subunit and is found in the bacterium G. sulfurreducens. They found that the opening process was mediated by a ligand that contains adenine, such as NAD+ or ADP, and the channel was closed by the presence of calcium ions. And by determining multiple crystal structures, Kong et al. were able to understand, from a structural point of view, how these ligands regulate this channel, and to propose a gating mechanism that is distinct from the mechanisms that are known to control other potassium channels. DOI:http://dx.doi.org/10.7554/eLife.00184.002
Collapse
Affiliation(s)
- Chunguang Kong
- Department of Physiology , University of Texas Southwestern Medical Center , Dallas , United States
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Rothberg BS. The BK channel: a vital link between cellular calcium and electrical signaling. Protein Cell 2012; 3:883-92. [PMID: 22996175 PMCID: PMC4875380 DOI: 10.1007/s13238-012-2076-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/23/2012] [Indexed: 10/27/2022] Open
Abstract
Large-conductance Ca²⁺-activated K⁺ channels (BK channels) constitute an key physiological link between cellular Ca²⁺ signaling and electrical signaling at the plasma membrane. Thus these channels are critical to the control of action potential firing and neurotransmitter release in several types of neurons, as well as the dynamic control of smooth muscle tone in resistance arteries, airway, and bladder. Recent advances in our understanding of K⁺ channel structure and function have led to new insight toward the molecular mechanisms of opening and closing (gating) of these channels. Here we will focus on mechanisms of BK channel gating by Ca²⁺, transmembrane voltage, and auxiliary subunit proteins.
Collapse
Affiliation(s)
- Brad S Rothberg
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
50
|
Pantazis A, Olcese R. Relative transmembrane segment rearrangements during BK channel activation resolved by structurally assigned fluorophore-quencher pairing. ACTA ACUST UNITED AC 2012; 140:207-18. [PMID: 22802360 PMCID: PMC3409098 DOI: 10.1085/jgp.201210807] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Voltage-activated proteins can sense, and respond to, changes in the electric field pervading the cell membrane by virtue of a transmembrane helix bundle, the voltage-sensing domain (VSD). Canonical VSDs consist of four transmembrane helices (S1-S4) of which S4 is considered a principal component because it possesses charged residues immersed in the electric field. Membrane depolarization compels the charges, and by extension S4, to rearrange with respect to the field. The VSD of large-conductance voltage- and Ca-activated K(+) (BK) channels exhibits two salient inconsistencies from the canonical VSD model: (1) the BK channel VSD possesses an additional nonconserved transmembrane helix (S0); and (2) it exhibits a "decentralized" distribution of voltage-sensing charges, in helices S2 and S3, in addition to S4. Considering these unique features, the voltage-dependent rearrangements of the BK VSD could differ significantly from the standard model of VSD operation. To understand the mode of operation of this unique VSD, we have optically tracked the relative motions of the BK VSD transmembrane helices during activation, by manipulating the quenching environment of site-directed fluorescent labels with native and introduced Trp residues. Having previously reported that S0 and S4 diverge during activation, in this work we demonstrate that S4 also diverges from S1 and S2, whereas S2, compelled by its voltage-sensing charged residues, moves closer to S1. This information contributes spatial constraints for understanding the BK channel voltage-sensing process, revealing the structural rearrangements in a non-canonical VSD.
Collapse
Affiliation(s)
- Antonios Pantazis
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90075, USA
| | | |
Collapse
|