1
|
Hein TW, Razavi HM, Xu X, Somvanshi S, Muthuchamy M, Kuo L. Activation of Smooth Muscle K ir2.1 Channels and Na +/K +-ATPase Mediates Dilation of Porcine Coronary Arterioles at Physiological Levels of Potassium. Int J Mol Sci 2025; 26:2654. [PMID: 40141296 PMCID: PMC11941845 DOI: 10.3390/ijms26062654] [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: 01/01/2025] [Revised: 01/31/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic stress on the heart can cause dilation of coronary arterioles for blood flow recruitment. Although potassium ions (K+) released from the myocardium are a major mediator for this response, the underlying signaling pathways for vasodilation are incompletely understood. Herein, the roles of smooth muscle inward-rectifier K+ channel subtype 2.1 (Kir2.1) and Na+/K+-ATPase were examined. Porcine coronary arterioles were isolated, cannulated, and pressurized for vasomotor study. Vessels developed basal tone and dilated concentration-dependently to extraluminal K+ from 7 to 20 mM. Higher K+ concentrations (25-40 mM) caused graded vasoconstriction. Vasodilation to K+ (10 mM) was not altered by endothelial removal, and blockade of ATP-sensitive K+ channels, voltage-sensitive K+ channels, or calcium-activated K+ channels did not affect K+-induced vasodilation. However, sustained but not abrupt transient vasodilation to K+ was reduced by the nonspecific Kir channel inhibitor Ba2+ or Kir2.1 channel blocker chloroethylclonidine. The Na+/K+-ATPase inhibitor ouabain attenuated K+-elicited vasodilation, and ouabain with Ba2+ abolished the response. Transfection of arterioles with Kir2.1 antisense oligonucleotides abolished sustained but not transient dilation. It is concluded that extraluminal K+ elevation within the physiological range induces initial transient dilation of porcine coronary arterioles by activating smooth muscle Na+/K+-ATPase and sustained dilation via smooth muscle Kir2.1 channels.
Collapse
Affiliation(s)
- Travis W. Hein
- Department of Medical Physiology, Cardiovascular Research Institute, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA (M.M.); (L.K.)
| | | | | | | | | | | |
Collapse
|
2
|
Connolly JG, Plant LD. SUMO Regulation of Ion Channels in Health and Disease. Physiology (Bethesda) 2025; 40:0. [PMID: 39499247 DOI: 10.1152/physiol.00034.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/07/2024] Open
Abstract
The small ubiquitin-like modifier (SUMO) protein pathway governs a panoply of vital biological processes including cell death, proliferation, differentiation, metabolism, and signal transduction by diversifying the functions, half-lives, and partnerships of target proteins in situ. More recently, SUMOylation has emerged as a key regulator of ion homeostasis and excitability across multiple tissues due to the regulation of a plethora of ion channels expressed in a range of tissue subtypes. Altogether, the balance of SUMOylation states among relevant ion channels can result in graded biophysical effects that tune excitability and contribute to a range of disease states including cardiac arrhythmia, epilepsy, pain transmission, and inflammation. Here, we consolidate these concepts by focusing on the role of ion channel SUMOylation in the central nervous system, peripheral nervous system, and cardiovascular system. In addition, we review what is known about the enigmatic factors that regulate the SUMO pathway and consider the emerging role of small molecule SUMO modulators as potential therapeutics in a range of diseases.
Collapse
Affiliation(s)
- Jenna G Connolly
- Department of Pharmaceutical Sciences and the Center for Drug Discovery, The School of Pharmacy and Pharmaceutical SciencesBouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, United States
| | - Leigh D Plant
- Department of Pharmaceutical Sciences and the Center for Drug Discovery, The School of Pharmacy and Pharmaceutical SciencesBouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, United States
| |
Collapse
|
3
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
4
|
Mayar S, Borbuliak M, Zoumpoulakis A, Bouceba T, Labonté MM, Ahrari A, Sinniah N, Memarpoor-Yazdi M, Vénien-Bryan C, Tieleman DP, D'Avanzo N. Endocannabinoid regulation of inward rectifier potassium (Kir) channels. Front Pharmacol 2024; 15:1439767. [PMID: 39253376 PMCID: PMC11381239 DOI: 10.3389/fphar.2024.1439767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
The inward rectifier potassium channel Kir2.1 (KCNJ2) is an important regulator of resting membrane potential in both excitable and non-excitable cells. The functions of Kir2.1 channels are dependent on their lipid environment, including the availability of PI(4,5)P2, secondary anionic lipids, cholesterol and long-chain fatty acids acyl coenzyme A (LC-CoA). Endocannabinoids are a class of lipids that are naturally expressed in a variety of cells, including cardiac, neuronal, and immune cells. While these lipids are identified as ligands for cannabinoid receptors there is a growing body of evidence that they can directly regulate the function of numerous ion channels independently of CBRs. Here we examine the effects of a panel of endocannabinoids on Kir2.1 function and demonstrate that a subset of endocannabinoids can alter Kir2.1 conductance to varying degrees independently of CBRs. Using computational and Surface plasmon resonance analysis, endocannabinoid regulation of Kir2.1 channels appears to be the result of altered membrane properties, rather than through direct protein-lipid interactions. Furthermore, differences in endocannabinoid effects on Kir4.1 and Kir7.1 channels, indicating that endocannabinoid regulation is not conserved among Kir family members. These findings may have broader implications on the function of cardiac, neuronal and/or immune cells.
Collapse
Affiliation(s)
- Sultan Mayar
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| | - Mariia Borbuliak
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, 2500 University Drive NW, Calgary, AB, Canada
| | - Andreas Zoumpoulakis
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC), UMR7590, Sorbonne Université, Centre national de la recherche scientifique (CNRS), MNHN, Paris, France
| | - Tahar Bouceba
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Protein Engineering Platform, Molecular Interaction Service, Paris, France
| | - Madeleine M Labonté
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| | - Ameneh Ahrari
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| | - Niveny Sinniah
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| | - Mina Memarpoor-Yazdi
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| | - Catherine Vénien-Bryan
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC), UMR7590, Sorbonne Université, Centre national de la recherche scientifique (CNRS), MNHN, Paris, France
| | - D Peter Tieleman
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, 2500 University Drive NW, Calgary, AB, Canada
| | - Nazzareno D'Avanzo
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
5
|
Lee BH, De Jesús Pérez JJ, Moiseenkova-Bell V, Rohacs T. Structural basis of the activation of TRPV5 channels by long-chain acyl-Coenzyme-A. Nat Commun 2023; 14:5883. [PMID: 37735536 PMCID: PMC10514044 DOI: 10.1038/s41467-023-41577-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 09/10/2023] [Indexed: 09/23/2023] Open
Abstract
Long-chain acyl-coenzyme A (LC-CoA) is a crucial metabolic intermediate that plays important cellular regulatory roles, including activation and inhibition of ion channels. The structural basis of ion channel regulation by LC-CoA is not known. Transient receptor potential vanilloid 5 and 6 (TRPV5 and TRPV6) are epithelial calcium-selective ion channels. Here, we demonstrate that LC-CoA activates TRPV5 and TRPV6 in inside-out patches, and both exogenously supplied and endogenously produced LC-CoA can substitute for the natural ligand phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) in maintaining channel activity in intact cells. Utilizing cryo-electron microscopy, we determined the structure of LC-CoA-bound TRPV5, revealing an open configuration with LC-CoA occupying the same binding site as PI(4,5)P2 in previous studies. This is consistent with our finding that PI(4,5)P2 could not further activate the channels in the presence of LC-CoA. Our data provide molecular insights into ion channel regulation by a metabolic signaling molecule.
Collapse
Affiliation(s)
- Bo-Hyun Lee
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, USA
- Department of Physiology, Gyeongsang National University Medical School, Jinju, Korea
| | - José J De Jesús Pérez
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Vera Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
6
|
Gada KD, Kamuene JM, Chandrashekar A, Kissell RC, Yauch AK, Plant LD. PI(4,5)P2 regulates the gating of NaV1.4 channels. J Gen Physiol 2023; 155:e202213255. [PMID: 37043561 PMCID: PMC10103707 DOI: 10.1085/jgp.202213255] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/22/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Voltage-gated sodium (NaV) channels are densely expressed in most excitable cells and activate in response to depolarization, causing a rapid influx of Na+ ions that initiates the action potential. The voltage-dependent activation of NaV channels is followed almost instantaneously by fast inactivation, setting the refractory period of excitable tissues. The gating cycle of NaV channels is subject to tight regulation, with perturbations leading to a range of pathophysiological states. The gating properties of most ion channels are regulated by the membrane phospholipid, phosphatidylinositol (4,5) bisphosphate (PI(4,5)P2). However, it is not known whether PI(4,5)P2 modulates the activity of NaV channels. Here, we utilize optogenetics to activate specific, membrane-associated phosphoinositide (PI)-phosphatases that dephosphorylate PI(4,5)P2 while simultaneously recording NaV1.4 channel currents. We show that dephosphorylating PI(4,5)P2 left-shifts the voltage-dependent gating of NaV1.4 to more hyperpolarized membrane potentials, augments the late current that persists after fast inactivation, and speeds the rate at which channels recover from fast inactivation. These effects are opposed by exogenous diC8PI(4,5)P2. We provide evidence that PI(4,5)P2 is a negative regulator that tunes the gating behavior of NaV1.4 channels.
Collapse
Affiliation(s)
- Kirin D. Gada
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Jordie M. Kamuene
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Aishwarya Chandrashekar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - R. Charles Kissell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Anne K. Yauch
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Leigh D. Plant
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| |
Collapse
|
7
|
Moreno-Manuel AI, Gutiérrez LK, Vera-Pedrosa ML, Cruz FM, Bermúdez-Jiménez FJ, Martínez-Carrascoso I, Sánchez-Pérez P, Macías Á, Jalife J. Molecular stratification of arrhythmogenic mechanisms in the Andersen Tawil syndrome. Cardiovasc Res 2023; 119:919-932. [PMID: 35892314 PMCID: PMC10153646 DOI: 10.1093/cvr/cvac118] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/12/2022] Open
Abstract
Andersen-Tawil syndrome (ATS) is a rare inheritable disease associated with loss-of-function mutations in KCNJ2, the gene coding the strong inward rectifier potassium channel Kir2.1, which forms an essential membrane protein controlling cardiac excitability. ATS is usually marked by a triad of periodic paralysis, life-threatening cardiac arrhythmias and dysmorphic features, but its expression is variable and not all patients with a phenotype linked to ATS have a known genetic alteration. The mechanisms underlying this arrhythmogenic syndrome are poorly understood. Knowing such mechanisms would be essential to distinguish ATS from other channelopathies with overlapping phenotypes and to develop individualized therapies. For example, the recently suggested role of Kir2.1 as a countercurrent to sarcoplasmic calcium reuptake might explain the arrhythmogenic mechanisms of ATS and its overlap with catecholaminergic polymorphic ventricular tachycardia. Here we summarize current knowledge on the mechanisms of arrhythmias leading to sudden cardiac death in ATS. We first provide an overview of the syndrome and its pathophysiology, from the patient's bedside to the protein and discuss the role of essential regulators and interactors that could play a role in cases of ATS. The review highlights novel ideas related to some post-translational channel interactions with partner proteins that might help define the molecular bases of the arrhythmia phenotype. We then propose a new all-embracing classification of the currently known ATS loss-of-function mutations according to their position in the Kir2.1 channel structure and their functional implications. We also discuss specific ATS pathogenic variants, their clinical manifestations, and treatment stratification. The goal is to provide a deeper mechanistic understanding of the syndrome toward the development of novel targets and personalized treatment strategies.
Collapse
Affiliation(s)
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | | | - Francisco Miguel Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Francisco José Bermúdez-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
- Departamento de Cardiología, Hospital Virgen de las Nieves, GranadaSpain
| | | | - Patricia Sánchez-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Stojilkovic SS, Balla T. PI(4,5)P2-dependent and -independent roles of PI4P in the control of hormone secretion by pituitary cells. Front Endocrinol (Lausanne) 2023; 14:1118744. [PMID: 36777340 PMCID: PMC9911653 DOI: 10.3389/fendo.2023.1118744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/28/2023] Open
Abstract
Plasma membrane and organelle membranes are home to seven phosphoinositides, an important class of low-abundance anionic signaling lipids that contribute to cellular functions by recruiting cytoplasmic proteins or interacting with the cytoplasmic domains of membrane proteins. Here, we briefly review the functions of three phosphoinositides, PI4P, PI(4,5)P2, and PI(3,4,5)P3, in cellular signaling and exocytosis, focusing on hormone-producing pituitary cells. PI(4,5)P2, acting as a substrate for phospholipase C, plays a key role in the control of pituitary cell functions, including hormone synthesis and secretion. PI(4,5)P2 also acts as a substrate for class I PI3-kinases, leading to the generation of two intracellular messengers, PI(3,4,5)P3 and PI(3,4)P2, which act through their intracellular effectors, including Akt. PI(4,5)P2 can also influence the release of pituitary hormones acting as an intact lipid to regulate ion channel gating and concomitant calcium signaling, as well as the exocytic pathway. Recent findings also show that PI4P is not only a precursor of PI(4,5)P2, but also a key signaling molecule in many cell types, including pituitary cells, where it controls hormone secretion in a PI(4,5)P2-independent manner.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Stanko S. Stojilkovic,
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
9
|
Vaithianathan T, Schneider EH, Bukiya AN, Dopico AM. Cholesterol and PIP 2 Modulation of BK Ca Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:217-243. [PMID: 36988883 PMCID: PMC10683925 DOI: 10.1007/978-3-031-21547-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Ca2+/voltage-gated, large conductance K+ channels (BKCa) are formed by homotetrameric association of α (slo1) subunits. Their activity, however, is suited to tissue-specific physiology largely due to their association with regulatory subunits (β and γ types), chaperone proteins, localized signaling, and the channel's lipid microenvironment. PIP2 and cholesterol can modulate BKCa activity independently of downstream signaling, yet activating Ca2+i levels and regulatory subunits control ligand action. At physiological Ca2+i and voltages, cholesterol and PIP2 reduce and increase slo1 channel activity, respectively. Moreover, slo1 proteins provide sites that seem to recognize cholesterol and PIP2: seven CRAC motifs in the slo1 cytosolic tail and a string of positively charged residues (Arg329, Lys330, Lys331) immediately after S6, respectively. A model that could explain the modulation of BKCa activity by cholesterol and/or PIP2 is hypothesized. The roles of additional sites, whether in slo1 or BKCa regulatory subunits, for PIP2 and/or cholesterol to modulate BKCa function are also discussed.
Collapse
Affiliation(s)
- Thirumalini Vaithianathan
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elizabeth H Schneider
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anna N Bukiya
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Alex M Dopico
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
10
|
Bukiya AN, Rosenhouse-Dantsker A. From Crosstalk to Synergism: The Combined Effect of Cholesterol and PI(4,5)P 2 on Inwardly Rectifying Potassium Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:169-191. [PMID: 36988881 DOI: 10.1007/978-3-031-21547-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Inwardly rectifying potassium (Kir) channels are integral membrane proteins that control the flux of potassium ions across cell membranes and regulate membrane permeability. All eukaryotic Kir channels require the membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) for activation. In recent years, it has become evident that the function of many members of this family of channels is also mediated by another essential lipid-cholesterol. Here, we focus on members of the Kir2 and Kir3 subfamilies and their modulation by these two key lipids. We discuss how PI(4,5)P2 and cholesterol bind to Kir2 and Kir3 channels and how they affect channel activity. We also discuss the accumulating evidence indicating that there is interplay between PI(4,5)P2 and cholesterol in the modulation of Kir2 and Kir3 channels. In particular, we review the crosstalk between PI(4,5)P2 and cholesterol in the modulation of the ubiquitously expressed Kir2.1 channel and the synergy between these two lipids in the modulation of the Kir3.4 channel, which is primarily expressed in the heart. Additionally, we demonstrate that there is also synergy in the modulation of Kir3.2 channels, which are expressed in the brain. These observations suggest that alterations in the relative levels PI(4,5)P2 and cholesterol may fine-tune Kir channel activity.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | | |
Collapse
|
11
|
Driggers CM, Shyng SL. Mechanistic insights on KATP channel regulation from cryo-EM structures. J Gen Physiol 2022; 155:213723. [PMID: 36441147 PMCID: PMC9700523 DOI: 10.1085/jgp.202113046] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Gated by intracellular ATP and ADP, ATP-sensitive potassium (KATP) channels couple cell energetics with membrane excitability in many cell types, enabling them to control a wide range of physiological processes based on metabolic demands. The KATP channel is a complex of four potassium channel subunits from the Kir channel family, Kir6.1 or Kir6.2, and four sulfonylurea receptor subunits, SUR1, SUR2A, or SUR2B, from the ATP-binding cassette (ABC) transporter family. Dysfunction of KATP channels underlies several human diseases. The importance of these channels in human health and disease has made them attractive drug targets. How the channel subunits interact with one another and how the ligands interact with the channel to regulate channel activity have been long-standing questions in the field. In the past 5 yr, a steady stream of high-resolution KATP channel structures has been published using single-particle cryo-electron microscopy (cryo-EM). Here, we review the advances these structures bring to our understanding of channel regulation by physiological and pharmacological ligands.
Collapse
Affiliation(s)
- Camden M. Driggers
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR,Correspondence to Show-Ling Shyng:
| |
Collapse
|
12
|
Xu Y, Yang Y, Chandrashekar A, Gada KD, Masotti M, Baggetta AM, Connolly JG, Kawano T, Plant LD. Hypoxia inhibits the cardiac I K1 current through SUMO targeting Kir2.1 activation by PIP 2. iScience 2022; 25:104969. [PMID: 36060074 PMCID: PMC9437851 DOI: 10.1016/j.isci.2022.104969] [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: 10/13/2021] [Revised: 05/07/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide. Most deaths are sudden and occur secondary to the occlusion of coronary arteries resulting in a rapid decrease in cellular oxygen levels. Acute hypoxia is proarrhythmic, leading to disordered electrical signals, conduction block, and uncoordinated beating of the myocardium. Although acute hypoxia is recognized to perturb the electrophysiology of heart muscle, the mechanistic basis for the effect has remained elusive, hampering the development of targeted therapeutic interventions. Here, we show that acute hypoxia activates the redox-sensitive SUMO pathway in cardiomyocytes, causing rapid inhibition of the inward-rectifying K+ channel, Kir2.1. We find that SUMOylation decreases the activation of Kir2.1 channels by the membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2). These data provide a mechanistic basis for the proarrhythmic effects of acute hypoxia and offer a framework for understanding the central role of PIP2 in mediating the sequelae of hypoxia and SUMOylation in cardiovascular disease.
Collapse
Affiliation(s)
- Yu Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Yuchen Yang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Aishwarya Chandrashekar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Kirin D. Gada
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Meghan Masotti
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Austin M. Baggetta
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Jenna G. Connolly
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Takeharu Kawano
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Leigh D. Plant
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| |
Collapse
|
13
|
Sancho M, Fletcher J, Welsh DG. Inward Rectifier Potassium Channels: Membrane Lipid-Dependent Mechanosensitive Gates in Brain Vascular Cells. Front Cardiovasc Med 2022; 9:869481. [PMID: 35419431 PMCID: PMC8995785 DOI: 10.3389/fcvm.2022.869481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral arteries contain two primary and interacting cell types, smooth muscle (SMCs) and endothelial cells (ECs), which are each capable of sensing particular hemodynamic forces to set basal tone and brain perfusion. These biomechanical stimuli help confer tone within arterial networks upon which local neurovascular stimuli function. Tone development is intimately tied to arterial membrane potential (VM) and changes in intracellular [Ca2+] driven by voltage-gated Ca2+ channels (VGCCs). Arterial VM is in turn set by the dynamic interplay among ion channel species, the strongly inward rectifying K+ (Kir) channel being of special interest. Kir2 channels possess a unique biophysical signature in that they strongly rectify, display negative slope conductance, respond to elevated extracellular K+ and are blocked by micromolar Ba2+. While functional Kir2 channels are expressed in both smooth muscle and endothelium, they lack classic regulatory control, thus are often viewed as a simple background conductance. Recent literature has provided new insight, with two membrane lipids, phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol, noted to (1) stabilize Kir2 channels in a preferred open or closed state, respectively, and (2) confer, in association with the cytoskeleton, caveolin-1 (Cav1) and syntrophin, hemodynamic sensitivity. It is these aspects of vascular Kir2 channels that will be the primary focus of this review.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Maria Sancho,
| | - Jacob Fletcher
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Donald G. Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Donald G. Welsh,
| |
Collapse
|
14
|
Qiao P, Schrecke S, Walker T, McCabe JW, Lyu J, Zhu Y, Zhang T, Kumar S, Clemmer D, Russell DH, Laganowsky A. Entropy in the Molecular Recognition of Membrane Protein-Lipid Interactions. J Phys Chem Lett 2021; 12:12218-12224. [PMID: 34928154 PMCID: PMC8905501 DOI: 10.1021/acs.jpclett.1c03750] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Understanding the molecular driving forces that underlie membrane protein-lipid interactions requires the characterization of their binding thermodynamics. Here, we employ variable-temperature native mass spectrometry to determine the thermodynamics of lipid binding events to the human G-protein-gated inward rectifier potassium channel, Kir3.2. The channel displays distinct thermodynamic strategies to engage phosphatidylinositol (PI) and phosphorylated forms thereof. The addition of a 4'-phosphate to PI results in an increase in favorable entropy. PI with two or more phosphates exhibits more complex binding, where lipids appear to bind two nonidentical sites on Kir3.2. Remarkably, the interaction of 4,5-bisphosphate PI with Kir3.2 is solely driven by a large, favorable change in entropy. Installment of a 3'-phosphate to PI(4,5)P2 results in an altered thermodynamic strategy. The acyl chain of the lipid has a marked impact on binding thermodynamics and, in some cases, enthalpy becomes favorable.
Collapse
Affiliation(s)
- Pei Qiao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Thomas Walker
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Jacob W McCabe
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Smriti Kumar
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - David Clemmer
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - David H Russell
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
15
|
Qiao P, Schrecke S, Lyu J, Zhu Y, Zhang T, Benavides A, Laganowsky A. Insight into the Phospholipid-Binding Preferences of Kir3.4. Biochemistry 2021; 60:3813-3821. [PMID: 34846128 DOI: 10.1021/acs.biochem.1c00615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The G-protein-gated inwardly rectifying potassium channel 4 (Kir3.4) subunit forms functional tetramers. Previous studies have established that phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is required for Kir3.4 function. However, the binding preferences of Kir3.4 for the headgroup and acyl chains of phosphorylated phosphatidylinositides (PIPs) and other lipids are not well understood. Here, the interactions between full-length, human Kir3.4 and lipids are characterized using native mass spectrometry (MS) in conjunction with a soluble fluorescent lipid-binding assay. Kir3.4 displays binding preferences for PIPs, and, in some cases, the degree of binding is influenced by the type of acyl chains. The interactions between Kir3.4 and PIPs are weaker in comparison to full-length, human Kir3.2. The binding of PI(4,5)P2 modified with a fluorophore to Kir3.2 can be enhanced by other lipids, such as phosphatidylcholine. Introduction of S143T, a mutation that enhances Kir3.4 activity, results in an overall reduction in the channel binding PIPs. In contrast, the D223N mutant of Kir3.4 that mimics the sodium-bound state exhibited stronger binding for PI(4,5)P2, particularly for those with 18:0-20:4 acyl chains. Taken together, these results provide additional insight into the interaction between Kir3.4 and lipids that are important for channel function.
Collapse
Affiliation(s)
- Pei Qiao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Amanda Benavides
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
16
|
Crotti L, Odening KE, Sanguinetti MC. Heritable arrhythmias associated with abnormal function of cardiac potassium channels. Cardiovasc Res 2021; 116:1542-1556. [PMID: 32227190 DOI: 10.1093/cvr/cvaa068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes express a surprisingly large number of potassium channel types. The primary physiological functions of the currents conducted by these channels are to maintain the resting membrane potential and mediate action potential repolarization under basal conditions and in response to changes in the concentrations of intracellular sodium, calcium, and ATP/ADP. Here, we review the diversity and functional roles of cardiac potassium channels under normal conditions and how heritable mutations in the genes encoding these channels can lead to distinct arrhythmias. We briefly review atrial fibrillation and J-wave syndromes. For long and short QT syndromes, we describe their genetic basis, clinical manifestation, risk stratification, traditional and novel therapeutic approaches, as well as insights into disease mechanisms provided by animal and cellular models.
Collapse
Affiliation(s)
- Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, San Luca Hospital, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Institute of Experimental Cardiovascular Medicine, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Department of Cardiology, Translational Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Michael C Sanguinetti
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
17
|
Pipatpolkai T, Quetschlich D, Stansfeld PJ. From Bench to Biomolecular Simulation: Phospholipid Modulation of Potassium Channels. J Mol Biol 2021; 433:167105. [PMID: 34139216 PMCID: PMC8361781 DOI: 10.1016/j.jmb.2021.167105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/05/2022]
Abstract
Potassium (K+) ion channels are crucial in numerous cellular processes as they hyperpolarise a cell through K+ conductance, returning a cell to its resting potential. K+ channel mutations result in multiple clinical complications such as arrhythmia, neonatal diabetes and migraines. Since 1995, the regulation of K+ channels by phospholipids has been heavily studied using a range of interdisciplinary methods such as cellular electrophysiology, structural biology and computational modelling. As a result, K+ channels are model proteins for the analysis of protein-lipid interactions. In this review, we will focus on the roles of lipids in the regulation of K+ channels, and how atomic-level structures, along with experimental techniques and molecular simulations, have helped guide our understanding of the importance of phospholipid interactions.
Collapse
Affiliation(s)
- Tanadet Pipatpolkai
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK; OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PT, UK
| | - Daniel Quetschlich
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Chemistry, South Parks Road, Oxford OX1 3QZ, UK
| | - Phillip J Stansfeld
- School of Life Sciences & Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
18
|
Abstract
Ion channel are embedded in the lipid bilayers of biological membranes. Membrane phospholipids constitute a barrier to ion movement, and they have been considered for a long time as a passive environment for channel proteins. Membrane phospholipids, however, do not only serve as a passive amphipathic environment, but they also modulate channel activity by direct specific lipid-protein interactions. Phosphoinositides are quantitatively minor components of biological membranes, and they play roles in many cellular functions, including membrane traffic, cellular signaling and cytoskeletal organization. Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is mainly found in the inner leaflet of the plasma membrane. Its role as a potential ion channel regulator was first appreciated over two decades ago and by now this lipid is a well-established cofactor or regulator of many different ion channels. The past two decades witnessed the steady development of techniques to study ion channel regulation by phosphoinositides with progress culminating in recent cryoEM structures that allowed visualization of how PI(4,5)P2 opens some ion channels. This chapter will provide an overview of the methods to study regulation by phosphoinositides, focusing on plasma membrane ion channels and PI(4,5)P2.
Collapse
|
19
|
Sancho M, Welsh DG. K IR channels in the microvasculature: Regulatory properties and the lipid-hemodynamic environment. CURRENT TOPICS IN MEMBRANES 2020; 85:227-259. [PMID: 32402641 DOI: 10.1016/bs.ctm.2020.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Basal tone and perfusion control is set in cerebral arteries by the sensing of pressure and flow, key hemodynamic stimuli. These forces establish a contractile foundation within arterial networks upon which local neurovascular stimuli operate. This fundamental process is intimately tied to arterial VM and the rise in cytosolic [Ca2+] by the graded opening of voltage-operated Ca2+ channels. Arterial VM is in turn controlled by a dynamic interaction among several resident ion channels, KIR being one of particular significance. As the name suggests, KIR displays strong inward rectification, retains a small outward component, potentiated by extracellular K+ and blocked by micromolar Ba2+. Cerebrovascular KIR is unique from other K+ currents as it is present in both smooth muscle and endothelium yet lacking in classical regulatory modulation. Such observations have fostered the view that KIR is nothing more than a background conductance, activated by extracellular K+ and which passively facilitates dilation. Recent work in cell model systems has; however, identified two membrane lipids, phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol, that interact with KIR2.x, to stabilize the channel in the preferred open or silent state, respectively. Translating this unique form of regulation, recent studies have demonstrated that specific lipid-protein interactions enable unique KIR populations to sense distinct hemodynamic stimuli and set basal tone. This review summarizes the current knowledge of vascular KIR channels and how the lipid and hemodynamic impact their activity.
Collapse
Affiliation(s)
- Maria Sancho
- Robarts Research Institute and the Department of Physiology & Pharmacology, University of Western Ontario, London, ON, Canada
| | - Donald G Welsh
- Robarts Research Institute and the Department of Physiology & Pharmacology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
20
|
Qiao P, Liu Y, Zhang T, Benavides A, Laganowsky A. Insight into the Selectivity of Kir3.2 toward Phosphatidylinositides. Biochemistry 2020; 59:2089-2099. [DOI: 10.1021/acs.biochem.0c00163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Pei Qiao
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Yang Liu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Amanda Benavides
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
21
|
Hammond GRV, Burke JE. Novel roles of phosphoinositides in signaling, lipid transport, and disease. Curr Opin Cell Biol 2020; 63:57-67. [PMID: 31972475 PMCID: PMC7247936 DOI: 10.1016/j.ceb.2019.12.007] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/22/2022]
Abstract
Phosphoinositides (PPIns) are lipid signaling molecules that act as master regulators of cellular signaling. Recent studies have revealed novel roles of PPIns in myriad cellular processes and multiple human diseases mediated by misregulation of PPIn signaling. This review will present a timely summary of recent discoveries in PPIn biology, specifically their role in regulating unexpected signaling pathways, modification of signaling outcomes downstream of integral membrane proteins, and novel roles in lipid transport. This has revealed new roles of PPIns in regulating membrane trafficking, immunity, cell polarity, and response to extracellular signals. A specific focus will be on novel opportunities to target PPIn metabolism for treatment of human diseases, including cancer, pathogen infection, developmental disorders, and immune disorders.
Collapse
Affiliation(s)
- Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, V8W 2Y2, Canada.
| |
Collapse
|
22
|
Fancher IS, Levitan I. Endothelial inwardly-rectifying K + channels as a key component of shear stress-induced mechanotransduction. CURRENT TOPICS IN MEMBRANES 2020; 85:59-88. [PMID: 32402645 DOI: 10.1016/bs.ctm.2020.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been recognized for decades that fluid shear stress plays a major role in vascular function. Acting on the endothelium shear stress induces vasorelaxation of resistance arteries and plays a major role in the propensity of the major arteries to atherosclerosis. Many elements of shear-induced signaling have been identified yet we are just beginning to decipher the roles that mechanosensitive ion channels may play in the signaling pathways initiated by shear stress. Endothelial inwardly-rectifying K+ channels were identified as potential primary mechanosensors in the late 1980s yet until our recent works, highlighted in the forthcoming chapter, the functional effect of a shear-activated K+ current was completely unknown. In this chapter, we present the physiological effects of shear stress in arteries in health and disease and highlight the most prevalent of today's investigated mechanosensitive ion channels. Ultimately, we focus on Kir2.1 channels and discuss in detail our findings regarding the downstream signaling events that are induced by shear-activated endothelial Kir2.1 channels. Most importantly, we examine our findings regarding hypercholesterolemia-induced inhibition of Kir channel shear-sensitivity and the impact on endothelial function in the context of flow (shear)-mediated vasodilation and atherosclerosis.
Collapse
Affiliation(s)
- Ibra S Fancher
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
23
|
How is the acyl chain composition of phosphoinositides created and does it matter? Biochem Soc Trans 2020; 47:1291-1305. [PMID: 31657437 PMCID: PMC6824679 DOI: 10.1042/bst20190205] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
The phosphoinositide (PIPn) family of signalling phospholipids are central regulators in membrane cell biology. Their varied functions are based on the phosphorylation pattern of their inositol ring, which can be recognized by selective binding domains in their effector proteins and be modified by a series of specific PIPn kinases and phosphatases, which control their interconversion in a spatial and temporal manner. Yet, a unique feature of PIPns remains largely unexplored: their unusually uniform acyl chain composition. Indeed, while most phospholipids present a range of molecular species comprising acyl chains of diverse length and saturation, PIPns in several organisms and tissues show the predominance of a single hydrophobic backbone, which in mammals is composed of arachidonoyl and stearoyl chains. Despite evolution having favoured this specific PIPn configuration, little is known regarding the mechanisms and functions behind it. In this review, we explore the metabolic pathways that could control the acyl chain composition of PIPns as well as the potential roles of this selective enrichment. While our understanding of this phenomenon has been constrained largely by the technical limitations in the methods traditionally employed in the PIPn field, we believe that the latest developments in PIPn analysis should shed light onto this old question.
Collapse
|
24
|
Duncan AL, Song W, Sansom MSP. Lipid-Dependent Regulation of Ion Channels and G Protein-Coupled Receptors: Insights from Structures and Simulations. Annu Rev Pharmacol Toxicol 2019; 60:31-50. [PMID: 31506010 DOI: 10.1146/annurev-pharmtox-010919-023411] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ion channels and G protein-coupled receptors (GPCRs) are regulated by lipids in their membrane environment. Structural studies combined with biophysical and molecular simulation investigations reveal interaction sites for specific lipids on membrane protein structures. For K channels, PIP2 plays a key role in regulating Kv and Kir channels. Likewise, several recent cryo-EM structures of TRP channels have revealed bound lipids, including PIP2 and cholesterol. Among the pentameric ligand-gated ion channel family, structural and biophysical studies suggest the M4 TM helix may act as a lipid sensor, e.g., forming part of the binding sites for neurosteroids on the GABAA receptor. Structures of GPCRs have revealed multiple cholesterol sites, which may modulate both receptor dynamics and receptor oligomerization. PIP2 also interacts with GPCRs and may modulate their interactions with G proteins. Overall, it is evident that multiple lipid binding sites exist on channels and receptors that modulate their function allosterically and are potential druggable sites.
Collapse
Affiliation(s)
- Anna L Duncan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom;
| | - Wanling Song
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom;
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom;
| |
Collapse
|
25
|
Le SC, Jia Z, Chen J, Yang H. Molecular basis of PIP 2-dependent regulation of the Ca 2+-activated chloride channel TMEM16A. Nat Commun 2019; 10:3769. [PMID: 31434906 PMCID: PMC6704070 DOI: 10.1038/s41467-019-11784-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/02/2019] [Indexed: 11/20/2022] Open
Abstract
The calcium-activated chloride channel (CaCC) TMEM16A plays crucial roles in regulating neuronal excitability, smooth muscle contraction, fluid secretion and gut motility. While opening of TMEM16A requires binding of intracellular Ca2+, prolonged Ca2+-dependent activation results in channel desensitization or rundown, the mechanism of which is unclear. Here we show that phosphatidylinositol (4,5)-bisphosphate (PIP2) regulates TMEM16A channel activation and desensitization via binding to a putative binding site at the cytosolic interface of transmembrane segments (TMs) 3-5. We further demonstrate that the ion-conducting pore of TMEM16A is constituted of two functionally distinct modules: a Ca2+-binding module formed by TMs 6-8 and a PIP2-binding regulatory module formed by TMs 3-5, which mediate channel activation and desensitization, respectively. PIP2 dissociation from the regulatory module results in ion-conducting pore collapse and subsequent channel desensitization. Our findings thus provide key insights into the mechanistic understanding of TMEM16 channel gating and lipid-dependent regulation.
Collapse
Affiliation(s)
- Son C Le
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Zhiguang Jia
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
26
|
Shimomura T, Kubo Y. Phosphoinositides modulate the voltage dependence of two-pore channel 3. J Gen Physiol 2019; 151:986-1006. [PMID: 31182502 PMCID: PMC6683669 DOI: 10.1085/jgp.201812285] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 02/03/2023] Open
Abstract
Two-pore channels, or two-pore Na+ channels (TPCs), contain two homologous domains, each containing a functional unit typical of voltage-dependent cation channels. Each domain is considered to be responsible for either phosphoinositide (PI) binding or voltage sensing. Among the three members of the TPC family, TPC1 and TPC2 are activated by PI(3,5)P2, while TPC3 has been thought not to be affected by any PIs. Here, we report that TPC3 is sensitive to PI(3,4)P2 and PI(3,5)P2, but not to PI(4,5)P2, and that the extremely slow increase in TPC3 currents induced by depolarization in Xenopus oocytes is due to the production of PI(3,4)P2 Similarly to TPC1, the cluster of basic amino acid residues in domain I is critical for PI sensitivity, but with a slight variation that may allow TPC3 to be sensitive to both PI(3,4)P2 and PI(3,5)P2 We also found that TPC3 has a unique PI-dependent modulation mechanism of voltage dependence, which is achieved by a specific bridging interaction between domain I and domain II. Taken together, these findings show that TPC3 is a unique member of the TPC family that senses PIs and displays a strong coupling between PI binding and voltage-dependent gating.
Collapse
Affiliation(s)
- Takushi Shimomura
- Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, Okazaki, Japan .,Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Japan
| |
Collapse
|
27
|
Sancho M, Fabris S, Hald BO, Brett SE, Sandow SL, Poepping TL, Welsh DG. Membrane Lipid-K
IR
2.x Channel Interactions Enable Hemodynamic Sensing in Cerebral Arteries. Arterioscler Thromb Vasc Biol 2019; 39:1072-1087. [DOI: 10.1161/atvbaha.119.312493] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective—
Inward rectifying K
+
(K
IR
) channels are present in cerebral arterial smooth muscle and endothelial cells, a tandem arrangement suggestive of a dynamic yet undiscovered role for this channel. This study defined whether distinct pools of cerebral arterial K
IR
channels were uniquely modulated by membrane lipids and hemodynamic stimuli.
Approach and Results—
A Ba
2+
-sensitive K
IR
current was isolated in smooth muscle and endothelial cells of rat cerebral arteries; molecular analyses subsequently confirmed K
IR
2.1/K
IR
2.2 mRNA and protein expression in both cells. Patch-clamp electrophysiology next demonstrated that each population of K
IR
channels was sensitive to key membrane lipids and hemodynamic stimuli. In this regard, endothelial K
IR
was sensitive to phosphatidylinositol 4,5-bisphosphate content, with depletion impairing the ability of laminar shear stress to activate this channel pool. In contrast, smooth muscle K
IR
was sensitive to membrane cholesterol content, with sequestration blocking the ability of pressure to inhibit channel activity. The idea that membrane lipids help confer shear stress and pressure sensitivity of K
IR
channels was confirmed in intact arteries using myography. Virtual models integrating structural/electrical observations reconceptualized K
IR
as a dynamic regulator of membrane potential working in concert with other currents to set basal tone across a range of shear stresses and intravascular pressures.
Conclusions—
The data show for the first time that specific membrane lipid-K
IR
interactions enable unique channel populations to sense hemodynamic stimuli and drive vasomotor responses to set basal perfusion in the cerebral circulation.
Collapse
Affiliation(s)
- Maria Sancho
- From the Department of Physiology and Pharmacology, Robarts Research Institute (M.S., S.F., S.E.B., D.G.W.), University of Western Ontario, London, Canada
| | - Sergio Fabris
- From the Department of Physiology and Pharmacology, Robarts Research Institute (M.S., S.F., S.E.B., D.G.W.), University of Western Ontario, London, Canada
| | - Bjorn O. Hald
- Department of Neuroscience, Translational Neurobiology, University of Copenhagen, Denmark (B.O.H.)
| | - Suzanne E. Brett
- From the Department of Physiology and Pharmacology, Robarts Research Institute (M.S., S.F., S.E.B., D.G.W.), University of Western Ontario, London, Canada
| | - Shaun L. Sandow
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia (S.L.S.)
| | - Tamie L. Poepping
- Department of Physics and Astronomy (T.L.P.), University of Western Ontario, London, Canada
| | - Donald G. Welsh
- From the Department of Physiology and Pharmacology, Robarts Research Institute (M.S., S.F., S.E.B., D.G.W.), University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology, University of Calgary, Alberta, Canada (D.G.W.)
| |
Collapse
|
28
|
Liu Y, LoCaste CE, Liu W, Poltash ML, Russell DH, Laganowsky A. Selective binding of a toxin and phosphatidylinositides to a mammalian potassium channel. Nat Commun 2019; 10:1352. [PMID: 30902995 PMCID: PMC6430785 DOI: 10.1038/s41467-019-09333-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 03/05/2019] [Indexed: 02/05/2023] Open
Abstract
G-protein-gated inward rectifying potassium channels (GIRKs) require Gβγ subunits and phosphorylated phosphatidylinositides (PIPs) for gating. Although studies have provided insight into these interactions, the mechanism of how these events are modulated by Gβγ and the binding affinity between PIPs and GIRKs remains poorly understood. Here, native ion mobility mass spectrometry is employed to directly monitor small molecule binding events to mouse GIRK2. GIRK2 binds the toxin tertiapin Q and PIPs selectively and with significantly higher affinity than other phospholipids. A mutation in GIRK2 that causes a rotation in the cytoplasmic domain, similarly to Gβγ-binding to the wild-type channel, revealed differences in the selectivity towards PIPs. More specifically, PIP isoforms known to weakly activate GIRKs have decreased binding affinity. Taken together, our results reveal selective small molecule binding and uncover a mechanism by which rotation of the cytoplasmic domain can modulate GIRK•PIP interactions.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, 77030, USA
| | - Catherine E LoCaste
- Department of Chemistry, Texas A&M University, College Station, TX, 77842, USA
| | - Wen Liu
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, 77030, USA
| | - Michael L Poltash
- Department of Chemistry, Texas A&M University, College Station, TX, 77842, USA
| | - David H Russell
- Department of Chemistry, Texas A&M University, College Station, TX, 77842, USA
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, TX, 77842, USA.
| |
Collapse
|
29
|
Choi S, Chen M, Cryns VL, Anderson RA. A nuclear phosphoinositide kinase complex regulates p53. Nat Cell Biol 2019; 21:462-475. [PMID: 30886346 DOI: 10.1038/s41556-019-0297-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/11/2019] [Indexed: 12/29/2022]
Abstract
The tumour suppressor p53 (encoded by TP53) protects the genome against cellular stress and is frequently mutated in cancer. Mutant p53 acquires gain-of-function oncogenic activities that are dependent on its enhanced stability. However, the mechanisms by which nuclear p53 is stabilized are poorly understood. Here, we demonstrate that the stability of stress-induced wild-type and mutant p53 is regulated by the type I phosphatidylinositol phosphate kinase (PIPKI-α (also known as PIP5K1A)) and its product phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2). Nuclear PIPKI-α binds to p53 upon stress, resulting in the production and association of PtdIns(4,5)P2 with p53. PtdIns(4,5)P2 binding promotes the interaction between p53 and the small heat shock proteins HSP27 (also known as HSPB1) and αB-crystallin (also known as HSPB5), which stabilize nuclear p53. Moreover, inhibition of PIPKI-α or PtdIns(4,5)P2 association results in p53 destabilization. Our results point to a previously unrecognized role of nuclear phosphoinositide signalling in regulating p53 stability and implicate this pathway as a promising therapeutic target in cancer.
Collapse
Affiliation(s)
- Suyong Choi
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Mo Chen
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
30
|
Understanding phosphoinositides: rare, dynamic, and essential membrane phospholipids. Biochem J 2019; 476:1-23. [PMID: 30617162 DOI: 10.1042/bcj20180022] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022]
Abstract
Polyphosphoinositides (PPIs) are essential phospholipids located in the cytoplasmic leaflet of eukaryotic cell membranes. Despite contributing only a small fraction to the bulk of cellular phospholipids, they make remarkable contributions to practically all aspects of a cell's life and death. They do so by recruiting cytoplasmic proteins/effectors or by interacting with cytoplasmic domains of membrane proteins at the membrane-cytoplasm interface to organize and mold organelle identity. The present study summarizes aspects of our current understanding concerning the metabolism, manipulation, measurement, and intimate roles these lipids play in regulating membrane homeostasis and vital cell signaling reactions in health and disease.
Collapse
|
31
|
Bozelli JC, Jennings W, Black S, Hou YH, Lameire D, Chatha P, Kimura T, Berno B, Khondker A, Rheinstädter MC, Epand RM. Membrane curvature allosterically regulates the phosphatidylinositol cycle, controlling its rate and acyl-chain composition of its lipid intermediates. J Biol Chem 2018; 293:17780-17791. [PMID: 30237168 DOI: 10.1074/jbc.ra118.005293] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/04/2018] [Indexed: 01/17/2023] Open
Abstract
Signaling events at membranes are often mediated by membrane lipid composition or membrane physical properties. These membrane properties could act either by favoring the membrane binding of downstream effectors or by modulating their activity. Several proteins can sense/generate membrane physical curvature (i.e. shape). However, the modulation of the activity of enzymes by a membrane's shape has not yet been reported. Here, using a cell-free assay with purified diacylglycerol kinase ϵ (DGKϵ) and liposomes, we studied the activity and acyl-chain specificity of an enzyme of the phosphatidylinositol (PI) cycle, DGKϵ. By systematically varying the model membrane lipid composition and physical properties, we found that DGKϵ has low activity and lacks acyl-chain specificity in locally flat membranes, regardless of the lipid composition. On the other hand, these enzyme properties were greatly enhanced in membrane structures with a negative Gaussian curvature. We also found that this is not a consequence of preferential binding of the enzyme to those structures, but rather is due to a curvature-mediated allosteric regulation of DGKϵ activity and acyl-chain specificity. Moreover, in a fine-tuned interplay between the enzyme and the membrane, DGKϵ favored the formation of structures with greater Gaussian curvature. DGKϵ does not bear a regulatory domain, and these findings reveal the importance of membrane curvature in regulating DGKϵ activity and acyl-chain specificity. Hence, this study highlights that a hierarchic coupling of membrane physical property and lipid composition synergistically regulates membrane signaling events. We propose that this regulatory mechanism of membrane-associated enzyme activity is likely more common than is currently appreciated.
Collapse
Affiliation(s)
- José Carlos Bozelli
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - William Jennings
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - Stephanie Black
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - Yu Heng Hou
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - Darius Lameire
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - Preet Chatha
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | - Tomohiro Kimura
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1
| | | | - Adree Khondker
- Physics and Astronomy; Origins Institute, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Maikel C Rheinstädter
- Physics and Astronomy; Origins Institute, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Richard M Epand
- From the Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, Ontario L8S 4K1; Departments of Chemistry.
| |
Collapse
|
32
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Han B, Zhang M, Sun P, Hou S. Capturing the Interaction Kinetics of an Ion Channel Protein with Small Molecules by the Bio-layer Interferometry Assay. J Vis Exp 2018. [PMID: 29578501 DOI: 10.3791/56846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The bio-layer interferometry (BLI) assay is a valuable tool for measuring protein-protein and protein-small molecule interactions. Here, we first describe the application of this novel label-free technique to study the interaction of human EAG1 (hEAG1) channel proteins with the small molecule PIP2. hEAG1 channel has been recognized as potential therapeutic target because of its aberrant overexpression in cancers and a few gain-of-function mutations involved in some types of neurological diseases. We purified hEAG1 channel proteins from a mammalian stable expression system and measured the interaction with PIP2 by BLI. The successful measurement of the kinetics of binding between hEAG1 protein and PIP2 demonstrates that the BLI assay is a potential high-throughput approach used for novel small-molecule ligand screening in ion channel pharmacology.
Collapse
Affiliation(s)
- Bo Han
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine
| | - Man Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Institute of Systems Biomedicine, Shanghai Jiao Tong University
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine
| | - Shangwei Hou
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine;
| |
Collapse
|
34
|
Ha J, Xu Y, Kawano T, Hendon T, Baki L, Garai S, Papapetropoulos A, Thakur GA, Plant LD, Logothetis DE. Hydrogen sulfide inhibits Kir2 and Kir3 channels by decreasing sensitivity to the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP 2). J Biol Chem 2018; 293:3546-3561. [PMID: 29317494 DOI: 10.1074/jbc.ra117.001679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/07/2018] [Indexed: 11/06/2022] Open
Abstract
Inwardly rectifying potassium (Kir) channels establish and regulate the resting membrane potential of excitable cells in the heart, brain, and other peripheral tissues. Phosphatidylinositol 4,5-bisphosphate (PIP2) is a key direct activator of ion channels, including Kir channels. The gasotransmitter carbon monoxide has been shown to regulate Kir channel activity by altering channel-PIP2 interactions. Here, we tested in two cellular models the effects and mechanism of action of another gasotransmitter, hydrogen sulfide (H2S), thought to play a key role in cellular responses under ischemic conditions. Direct administration of sodium hydrogen sulfide as an exogenous H2S source and expression of cystathionine γ-lyase, a key enzyme that produces endogenous H2S in specific brain tissues, resulted in comparable current inhibition of several Kir2 and Kir3 channels. This effect resulted from changes in channel-gating kinetics rather than in conductance or cell-surface localization. The extent of H2S regulation depended on the strength of the channel-PIP2 interactions. H2S regulation was attenuated when channel-PIP2 interactions were strengthened and was increased when channel-PIP2 interactions were weakened by depleting PIP2 levels. These H2S effects required specific cytoplasmic cysteine residues in Kir3.2 channels. Mutation of these residues abolished H2S inhibition, and reintroduction of specific cysteine residues back into the background of the cytoplasmic cysteine-lacking mutant rescued H2S inhibition. Molecular dynamics simulation experiments provided mechanistic insights into how potential sulfhydration of specific cysteine residues could lead to changes in channel-PIP2 interactions and channel gating.
Collapse
Affiliation(s)
- Junghoon Ha
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Yu Xu
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Takeharu Kawano
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Tyler Hendon
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Lia Baki
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Sumanta Garai
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Andreas Papapetropoulos
- the Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens 157 71, Greece, and.,the Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Leigh D Plant
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Diomedes E Logothetis
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, .,Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| |
Collapse
|
35
|
Cooper PE, McClenaghan C, Chen X, Stary-Weinzinger A, Nichols CG. Conserved functional consequences of disease-associated mutations in the slide helix of Kir6.1 and Kir6.2 subunits of the ATP-sensitive potassium channel. J Biol Chem 2017; 292:17387-17398. [PMID: 28842488 DOI: 10.1074/jbc.m117.804971] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/04/2017] [Indexed: 11/06/2022] Open
Abstract
Cantu syndrome (CS) is a condition characterized by a range of anatomical defects, including cardiomegaly, hyperflexibility of the joints, hypertrichosis, and craniofacial dysmorphology. CS is associated with multiple missense mutations in the genes encoding the regulatory sulfonylurea receptor 2 (SUR2) subunits of the ATP-sensitive K+ (KATP) channel as well as two mutations (V65M and C176S) in the Kir6.1 (KCNJ8) subunit. Previous analysis of leucine and alanine substitutions at the Val-65-equivalent site (Val-64) in Kir6.2 indicated no major effects on channel function. In this study, we characterized the effects of both valine-to-methionine and valine-to-leucine substitutions at this position in both Kir6.1 and Kir6.2 using ion flux and patch clamp techniques. We report that methionine substitution, but not leucine substitution, results in increased open state stability and hence significantly reduced ATP sensitivity and a marked increase of channel activity in the intact cell irrespective of the identity of the coassembled SUR subunit. Sulfonylurea inhibitors, such as glibenclamide, are potential therapies for CS. However, as a consequence of the increased open state stability, both Kir6.1(V65M) and Kir6.2(V64M) mutations essentially abolish high-affinity sensitivity to the KATP blocker glibenclamide in both intact cells and excised patches. This raises the possibility that, at least for some CS mutations, sulfonylurea therapy may not prove to be successful and highlights the need for detailed pharmacogenomic analyses of CS mutations.
Collapse
Affiliation(s)
- Paige E Cooper
- From the Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri 63110 and
| | - Conor McClenaghan
- From the Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri 63110 and
| | - Xingyu Chen
- Department of Pharmacology and Toxicology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Anna Stary-Weinzinger
- Department of Pharmacology and Toxicology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Colin G Nichols
- From the Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri 63110 and
| |
Collapse
|
36
|
Lacin E, Aryal P, Glaaser IW, Bodhinathan K, Tsai E, Marsh N, Tucker SJ, Sansom MSP, Slesinger PA. Dynamic role of the tether helix in PIP 2-dependent gating of a G protein-gated potassium channel. J Gen Physiol 2017; 149:799-811. [PMID: 28720589 PMCID: PMC5560777 DOI: 10.1085/jgp.201711801] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/06/2017] [Accepted: 06/21/2017] [Indexed: 01/21/2023] Open
Abstract
G protein–gated inwardly rectifying potassium (GIRK) channels are activated by the phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2). Using functional and computational experiments, Lacin et al. reveal that PIP2 interacts with the tether helix of the neuronal GIRK channel in a dynamic way. G protein–gated inwardly rectifying potassium (GIRK) channels control neuronal excitability in the brain and are implicated in several different neurological diseases. The anionic phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is an essential cofactor for GIRK channel gating, but the precise mechanism by which PIP2 opens GIRK channels remains poorly understood. Previous structural studies have revealed several highly conserved, positively charged residues in the “tether helix” (C-linker) that interact with the negatively charged PIP2. However, these crystal structures of neuronal GIRK channels in complex with PIP2 provide only snapshots of PIP2’s interaction with the channel and thus lack details about the gating transitions triggered by PIP2 binding. Here, our functional studies reveal that one of these conserved basic residues in GIRK2, Lys200 (6′K), supports a complex and dynamic interaction with PIP2. When Lys200 is mutated to an uncharged amino acid, it activates the channel by enhancing the interaction with PIP2. Atomistic molecular dynamic simulations of neuronal GIRK2 with the same 6′ substitution reveal an open GIRK2 channel with PIP2 molecules adopting novel positions. This dynamic interaction with PIP2 may explain the intrinsic low open probability of GIRK channels and the mechanism underlying activation by G protein Gβγ subunits and ethanol.
Collapse
Affiliation(s)
- Emre Lacin
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Prafulla Aryal
- Department of Biochemistry, University of Oxford, Oxford, England, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK
| | - Ian W Glaaser
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Eric Tsai
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nidaa Marsh
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stephen J Tucker
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK.,Department of Physics, University of Oxford, Oxford, England, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, England, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK
| | - Paul A Slesinger
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
37
|
Dual activation of neuronal G protein-gated inwardly rectifying potassium (GIRK) channels by cholesterol and alcohol. Sci Rep 2017; 7:4592. [PMID: 28676630 PMCID: PMC5496853 DOI: 10.1038/s41598-017-04681-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
Activation of G protein-gated inwardly rectifying potassium (GIRK) channels leads to a hyperpolarization of the neuron’s membrane potential, providing an important component of inhibition in the brain. In addition to the canonical G protein-activation pathway, GIRK channels are activated by small molecules but less is known about the underlying gating mechanisms. One drawback to previous studies has been the inability to control intrinsic and extrinsic factors. Here we used a reconstitution strategy with highly purified mammalian GIRK2 channels incorporated into liposomes and demonstrate that cholesterol or intoxicating concentrations of ethanol, i.e., >20 mM, each activate GIRK2 channels directly, in the absence of G proteins. Notably, both activators require the membrane phospholipid PIP2 but appear to interact independently with different regions of the channel. Elucidating the mechanisms underlying G protein-independent pathways of activating GIRK channels provides a unique strategy for developing new types of neuronal excitability modulators.
Collapse
|
38
|
York N, Halbach P, Chiu MA, Bird IM, Pillers DAM, Pattnaik BR. Oxytocin (OXT)-stimulated inhibition of Kir7.1 activity is through PIP 2-dependent Ca 2+ response of the oxytocin receptor in the retinal pigment epithelium in vitro. Cell Signal 2017; 37:93-102. [PMID: 28603013 DOI: 10.1016/j.cellsig.2017.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/28/2017] [Accepted: 06/05/2017] [Indexed: 01/26/2023]
Abstract
Oxytocin (OXT) is a neuropeptide that activates the oxytocin receptor (OXTR), a rhodopsin family G-protein coupled receptor. Our localization of OXTR to the retinal pigment epithelium (RPE), in close proximity to OXT in the adjacent photoreceptor neurons, leads us to propose that OXT plays an important role in RPE-retinal communication. An increase of RPE [Ca2+]i in response to OXT stimulation implies that the RPE may utilize oxytocinergic signaling as a mechanism by which it accomplishes some of its many roles. In this study, we used an established human RPE cell line, a HEK293 heterologous OXTR expression system, and pharmacological inhibitors of Ca2+ signaling to demonstrate that OXTR utilizes capacitative Ca2+ entry (CCE) mechanisms to sustain an increase in cytoplasmic Ca2+. These findings demonstrate how multiple functional outcomes of OXT-OXTR signaling could be integrated via a single pathway. In addition, the activated OXTR was able to inhibit the Kir7.1 channel, an important mediator of sub retinal waste transport and K+ homeostasis.
Collapse
Affiliation(s)
- Nathaniel York
- Endocrinology-Reproductive Physiology Program, The University of Wisconsin, Madison, WI 53715, United States; Division of Neonatology& Newborn Nursery, The University of Wisconsin, Madison, WI 53715, United States; Departments of Pediatrics, The University of Wisconsin, Madison, WI 53715, United States; The McPherson Eye Research Institute, The University of Wisconsin, Madison, WI 53715, United States
| | - Patrick Halbach
- Endocrinology-Reproductive Physiology Program, The University of Wisconsin, Madison, WI 53715, United States; Division of Neonatology& Newborn Nursery, The University of Wisconsin, Madison, WI 53715, United States; Departments of Pediatrics, The University of Wisconsin, Madison, WI 53715, United States; The McPherson Eye Research Institute, The University of Wisconsin, Madison, WI 53715, United States
| | - Michelle A Chiu
- Division of Neonatology& Newborn Nursery, The University of Wisconsin, Madison, WI 53715, United States; Departments of Pediatrics, The University of Wisconsin, Madison, WI 53715, United States; The McPherson Eye Research Institute, The University of Wisconsin, Madison, WI 53715, United States
| | - Ian M Bird
- Endocrinology-Reproductive Physiology Program, The University of Wisconsin, Madison, WI 53715, United States; Obstetrics & Gynecology, The University of Wisconsin, Madison, WI 53715, United States
| | - De-Ann M Pillers
- Division of Neonatology& Newborn Nursery, The University of Wisconsin, Madison, WI 53715, United States; Departments of Pediatrics, The University of Wisconsin, Madison, WI 53715, United States; Medical Genetics, The University of Wisconsin, Madison, WI 53715, United States; The McPherson Eye Research Institute, The University of Wisconsin, Madison, WI 53715, United States
| | - Bikash R Pattnaik
- Endocrinology-Reproductive Physiology Program, The University of Wisconsin, Madison, WI 53715, United States; Division of Neonatology& Newborn Nursery, The University of Wisconsin, Madison, WI 53715, United States; Departments of Pediatrics, The University of Wisconsin, Madison, WI 53715, United States; Ophthalmology &Visual Sciences, The University of Wisconsin, Madison, WI 53715, United States; The McPherson Eye Research Institute, The University of Wisconsin, Madison, WI 53715, United States.
| |
Collapse
|
39
|
Activation of the Ca 2+-sensing receptors increases currents through inward rectifier K + channels via activation of phosphatidylinositol 4-kinase. Pflugers Arch 2016; 468:1931-1943. [PMID: 27838849 PMCID: PMC5138266 DOI: 10.1007/s00424-016-1901-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/26/2016] [Accepted: 11/06/2016] [Indexed: 10/25/2022]
Abstract
Inward rectifier K+ channels are important for maintaining normal electrical function in many cell types. The proper function of these channels requires the presence of membrane phosphoinositide 4,5-bisphosphate (PIP2). Stimulation of the Ca2+-sensing receptor CaR, a pleiotropic G protein-coupled receptor, activates both Gq/11, which decreases PIP2, and phosphatidylinositol 4-kinase (PI-4-K), which, conversely, increases PIP2. How membrane PIP2 levels are regulated by CaR activation and whether these changes modulate inward rectifier K+ are unknown. In this study, we found that activation of CaR by the allosteric agonist, NPSR568, increased inward rectifier K+ current (I K1) in guinea pig ventricular myocytes and currents mediated by Kir2.1 channels exogenously expressed in HEK293T cells with a similar sensitivity. Moreover, using the fluorescent PIP2 reporter tubby-R332H-cYFP to monitor PIP2 levels, we found that CaR activation in HEK293T cells increased membrane PIP2 concentrations. Pharmacological studies showed that both phospholipase C (PLC) and PI-4-K are activated by CaR stimulation with the latter played a dominant role in regulating membrane PIP2 and, thus, Kir currents. These results provide the first direct evidence that CaR activation upregulates currents through inward rectifier K+ channels by accelerating PIP2 synthesis. The regulation of I K1 plays a critical role in the stability of the electrical properties of many excitable cells, including cardiac myocytes and neurons. Further, synthetic allosteric modulators that increase CaR activity have been used to treat hyperparathyroidism, and negative CaR modulators are of potential importance in the treatment of osteoporosis. Thus, our results provide further insight into the roles played by CaR in the cardiovascular system and are potentially valuable for heart disease treatment and drug safety.
Collapse
|
40
|
Nanoscale analysis reveals agonist-sensitive and heterogeneous pools of phosphatidylinositol 4-phosphate in the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:1298-305. [DOI: 10.1016/j.bbamem.2016.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 01/06/2023]
|
41
|
Human EAG channels are directly modulated by PIP2 as revealed by electrophysiological and optical interference investigations. Sci Rep 2016; 6:23417. [PMID: 27005320 PMCID: PMC4804213 DOI: 10.1038/srep23417] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/03/2016] [Indexed: 11/28/2022] Open
Abstract
Voltage-gated ether à go-go (EAG) K+ channels are expressed in various types of cancer cells and also in the central nervous system. Aberrant overactivation of human EAG1 (hEAG1) channels is associated with cancer and neuronal disorders such as Zimmermann-Laband and Temple-Baraitser syndromes. Although hEAG1 channels are recognized as potential therapeutic targets, regulation of their functional properties is only poorly understood. Here, we show that the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) is a potent inhibitory gating modifier of hEAG1 channels. PIP2 inhibits the channel activity by directly binding to a short N-terminal segment of the channel important for Ca2+/calmodulin (CaM) binding as evidenced by bio-layer interferometry measurements. Conversely, depletion of endogenous PIP2 either by serotonin-induced phospholipase C (PLC) activation or by a rapamycin-induced translocation system enhances the channel activity at physiological membrane potentials, suggesting that PIP2 exerts a tonic inhibitory influence. Our study, combining electrophysiological and direct binding assays, demonstrates that hEAG1 channels are subject to potent inhibitory modulation by multiple phospholipids and suggests that manipulations of the PIP2 signaling pathway may represent a strategy to treat hEAG1 channel-associated diseases.
Collapse
|
42
|
Tóth BI, Konrad M, Ghosh D, Mohr F, Halaszovich CR, Leitner MG, Vriens J, Oberwinkler J, Voets T. Regulation of the transient receptor potential channel TRPM3 by phosphoinositides. ACTA ACUST UNITED AC 2016; 146:51-63. [PMID: 26123194 PMCID: PMC4485019 DOI: 10.1085/jgp.201411339] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
TRPM3 is dynamically regulated by plasma membrane PI(4,5)P2 and related PIPs. The transient receptor potential (TRP) channel TRPM3 is a calcium-permeable cation channel activated by heat and by the neurosteroid pregnenolone sulfate (PregS). TRPM3 is highly expressed in sensory neurons, where it plays a key role in heat sensing and inflammatory hyperalgesia, and in pancreatic β cells, where its activation enhances glucose-induced insulin release. However, despite its functional importance, little is known about the cellular mechanisms that regulate TRPM3 activity. Here, we provide evidence for a dynamic regulation of TRPM3 by membrane phosphatidylinositol phosphates (PIPs). Phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) and ATP applied to the intracellular side of excised membrane patches promote recovery of TRPM3 from desensitization. The stimulatory effect of cytosolic ATP on TRPM3 reflects activation of phosphatidylinositol kinases (PI-Ks), leading to resynthesis of PIPs in the plasma membrane. Various PIPs directly enhance TRPM3 activity in cell-free inside-out patches, with a potency order PI(3,4,5)P3 > PI(3,5)P2 > PI(4,5)P2 ≈ PI(3,4)P2 >> PI(4)P. Conversely, TRPM3 activity is rapidly and reversibly inhibited by activation of phosphatases that remove the 5-phosphate from PIPs. Finally, we show that recombinant TRPM3, as well as the endogenous TRPM3 in insuloma cells, is rapidly and reversibly inhibited by activation of phospholipase C–coupled muscarinic acetylcholine receptors. Our results reveal basic cellular mechanisms whereby membrane receptors can regulate TRPM3 activity.
Collapse
Affiliation(s)
- Balázs I Tóth
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe) and Laboratory of Obstetrics and Experimental Gynaecology, KU Leuven, 3000 Leuven, Belgium
| | - Maik Konrad
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Debapriya Ghosh
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe) and Laboratory of Obstetrics and Experimental Gynaecology, KU Leuven, 3000 Leuven, Belgium
| | - Florian Mohr
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Christian R Halaszovich
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Michael G Leitner
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Joris Vriens
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe) and Laboratory of Obstetrics and Experimental Gynaecology, KU Leuven, 3000 Leuven, Belgium Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe) and Laboratory of Obstetrics and Experimental Gynaecology, KU Leuven, 3000 Leuven, Belgium
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe) and Laboratory of Obstetrics and Experimental Gynaecology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
43
|
Ufret-Vincenty CA, Klein RM, Collins MD, Rosasco MG, Martinez GQ, Gordon SE. Mechanism for phosphoinositide selectivity and activation of TRPV1 ion channels. ACTA ACUST UNITED AC 2016; 145:431-42. [PMID: 25918361 PMCID: PMC4411251 DOI: 10.1085/jgp.201511354] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphoinositides bind to a selective site in the proximal C-terminal region to regulate TRPV1. Although PI(4,5)P2 is believed to play an essential role in regulating the activity of numerous ion channels and transporters, the mechanisms by which it does so are unknown. Here, we used the ability of the TRPV1 ion channel to discriminate between PI(4,5)P2 and PI(4)P to localize the region of TRPV1 sequence that interacts directly with the phosphoinositide. We identified a point mutation in the proximal C-terminal region after the TRP box, R721A, that inverted the selectivity of TRPV1. Although the R721A mutation produced only a 30% increase in the EC50 for activation by PI(4,5)P2, it decreased the EC50 for activation by PI(4)P by more than two orders of magnitude. We used chemically induced and voltage-activated phosphatases to determine that PI(4)P continued to support TRPV1 activity even after depletion of PI(4,5)P2 from the plasma membrane. Our data cannot be explained by a purely electrostatic mechanism for interaction between the phosphoinositide and the protein, similar to that of the MARCKS (myristoylated alanine-rich C kinase substrate) effector domain or the EGF receptor. Rather, conversion of a PI(4,5)P2-selective channel to a PI(4)P-selective channel indicates that a structured phosphoinositide-binding site mediates the regulation of TRPV1 activity and that the amino acid at position 721 likely interacts directly with the moiety at the 5′ position of the phosphoinositide.
Collapse
Affiliation(s)
| | - Rebecca M Klein
- University of Washington School of Medicine, Seattle, WA 98195
| | | | - Mario G Rosasco
- University of Washington School of Medicine, Seattle, WA 98195
| | | | | |
Collapse
|
44
|
Requirement of Phosphoinositides Containing Stearic Acid To Control Cell Polarity. Mol Cell Biol 2015; 36:765-80. [PMID: 26711260 DOI: 10.1128/mcb.00843-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Phosphoinositides (PIPs) are present in very small amounts but are essential for cell signaling, morphogenesis, and polarity. By mass spectrometry, we demonstrated that some PIPs with stearic acyl chains were strongly disturbed in a psi1Δ Saccharomyces cerevisiae yeast strain deficient in the specific incorporation of a stearoyl chain at the sn-1 position of phosphatidylinositol. The absence of PIPs containing stearic acid induced disturbances in intracellular trafficking, although the total amount of PIPs was not diminished. Changes in PIPs also induced alterations in the budding pattern and defects in actin cytoskeleton organization (cables and patches). Moreover, when the PSI1 gene was impaired, a high proportion of cells with bipolar cortical actin patches that occurred concomitantly with the bipolar localization of Cdc42p was specifically found among diploid cells. This bipolar cortical actin phenotype, never previously described, was also detected in a bud9Δ/bud9Δ strain. Very interestingly, overexpression of PSI1 reversed this phenotype.
Collapse
|
45
|
Kim KS, Jang JH, Lin H, Choi SW, Kim HR, Shin DH, Nam JH, Zhang YH, Kim SJ. Rise and Fall of Kir2.2 Current by TLR4 Signaling in Human Monocytes: PKC-Dependent Trafficking and PI3K-Mediated PIP2 Decrease. THE JOURNAL OF IMMUNOLOGY 2015; 195:3345-54. [PMID: 26324774 DOI: 10.4049/jimmunol.1500056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 07/22/2015] [Indexed: 12/24/2022]
Abstract
LPSs are widely used to stimulate TLR4, but their effects on ion channels in immune cells are poorly known. In THP-1 cells and human blood monocytes treated with LPS, inwardly rectifying K(+) channel current (IKir,LPS) newly emerged at 1 h, peaked at 4 h (-119 ± 8.6 pA/pF), and decayed afterward (-32 ± 6.7 pA/pF at 24 h). Whereas both the Kir2.1 and Kir2.2 mRNAs and proteins were observed, single-channel conductance (38 pS) of IKir,LPS and small interfering RNA-induced knockdown commonly indicated Kir2.2 than Kir2.1. LPS-induced cytokine release and store-operated Ca(2+) entry were commonly decreased by ML-133, a Kir2 inhibitor. Immunoblot, confocal microscopy, and the effects of vesicular trafficking inhibitors commonly suggested plasma membrane translocation of Kir2.2 by LPS. Both IKir,LPS and membrane translocation of Kir2.2 were inhibited by GF109203X (protein kinase C [PKC] inhibitor) or by transfection with small interfering RNA-specific PKCε. Interestingly, pharmacological activation of PKC by PMA induced both Kir2.1 and Kir2.2 currents. The spontaneously decayed IKir,LPS at 24 h was recovered by PI3K inhibitors but further suppressed by an inhibitor of phosphatidylinositol(3,4,5)-trisphosphate (PIP3) phosphatase (phosphatase and tensin homolog). However, IKir,LPS at 24 h was not affected by Akt inhibitors, suggesting that the decreased phosphatidylinositol(4,5)-bisphosphate availability, that is, conversion into PIP3 by PI3K, per se accounts for the decay of IKir,LPS. Taken together, to our knowledge these data are the first demonstrations that IKir is newly induced by TLR4 stimulation via PKC-dependent membrane trafficking of Kir2.2, and that conversion of phosphatidylinositol(4,5)-bisphosphate to PIP3 modulates Kir2.2. The augmentation of Ca(2+) influx and cytokine release suggests a physiological role for Kir2.2 in TLR4-stimulated monocytes.
Collapse
Affiliation(s)
- Kyung Soo Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Ji Hyun Jang
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Haiyue Lin
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Seong Woo Choi
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Hang Rae Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Dong Hoon Shin
- Division of Natural Medical Sciences, College of Health Science, Chosun University, Gwangju 501-759, Republic of Korea; and
| | - Joo Hyun Nam
- Channelopathy Research Center, Dongguk University College of Medicine, Goyang 410-773, Republic of Korea
| | - Yin Hua Zhang
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Channelopathy Research Center, Dongguk University College of Medicine, Goyang 410-773, Republic of Korea
| |
Collapse
|
46
|
Glaaser IW, Slesinger PA. Structural Insights into GIRK Channel Function. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:117-60. [PMID: 26422984 DOI: 10.1016/bs.irn.2015.05.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G protein-gated inwardly rectifying potassium (GIRK; Kir3) channels, which are members of the large family of inwardly rectifying potassium channels (Kir1-Kir7), regulate excitability in the heart and brain. GIRK channels are activated following stimulation of G protein-coupled receptors that couple to the G(i/o) (pertussis toxin-sensitive) G proteins. GIRK channels, like all other Kir channels, possess an extrinsic mechanism of inward rectification involving intracellular Mg(2+) and polyamines that occlude the conduction pathway at membrane potentials positive to E(K). In the past 17 years, more than 20 high-resolution atomic structures containing GIRK channel cytoplasmic domains and transmembrane domains have been solved. These structures have provided valuable insights into the structural determinants of many of the properties common to all inward rectifiers, such as permeation and rectification, as well as revealing the structural bases for GIRK channel gating. In this chapter, we describe advances in our understanding of GIRK channel function based on recent high-resolution atomic structures of inwardly rectifying K(+) channels discussed in the context of classical structure-function experiments.
Collapse
Affiliation(s)
- Ian W Glaaser
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul A Slesinger
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
47
|
Identification of the Conformational transition pathway in PIP2 Opening Kir Channels. Sci Rep 2015; 5:11289. [PMID: 26063437 PMCID: PMC4462750 DOI: 10.1038/srep11289] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/29/2015] [Indexed: 11/08/2022] Open
Abstract
The gating of Kir channels depends critically on phosphatidylinositol 4,5-bisphosphate (PIP2), but the detailed mechanism by which PIP2 regulates Kir channels remains obscure. Here, we performed a series of Targeted molecular dynamics simulations on the full-length Kir2.1 channel and, for the first time, were able to achieve the transition from the closed to the open state. Our data show that with the upward motion of the cytoplasmic domain (CTD) the structure of the C-Linker changes from a loop to a helix. The twisting of the C-linker triggers the rotation of the CTD, which induces a small downward movement of the CTD and an upward motion of the slide helix toward the membrane that pulls the inner helix gate open. At the same time, the rotation of the CTD breaks the interaction between the CD- and G-loops thus releasing the G-loop. The G-loop then bounces away from the CD-loop, which leads to the opening of the G-loop gate and the full opening of the pore. We identified a series of interaction networks, between the N-terminus, CD loop, C linker and G loop one by one, which exquisitely regulates the global conformational changes during the opening of Kir channels by PIP2.
Collapse
|
48
|
Hille B, Dickson EJ, Kruse M, Vivas O, Suh BC. Phosphoinositides regulate ion channels. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1851:844-56. [PMID: 25241941 PMCID: PMC4364932 DOI: 10.1016/j.bbalip.2014.09.010] [Citation(s) in RCA: 472] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 11/29/2022]
Abstract
Phosphoinositides serve as signature motifs for different cellular membranes and often are required for the function of membrane proteins. Here, we summarize clear evidence supporting the concept that many ion channels are regulated by membrane phosphoinositides. We describe tools used to test their dependence on phosphoinositides, especially phosphatidylinositol 4,5-bisphosphate, and consider mechanisms and biological meanings of phosphoinositide regulation of ion channels. This lipid regulation can underlie changes of channel activity and electrical excitability in response to receptors. Since different intracellular membranes have different lipid compositions, the activity of ion channels still in transit towards their final destination membrane may be suppressed until they reach an optimal lipid environment. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
Affiliation(s)
- Bertil Hille
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA.
| | - Eamonn J Dickson
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA.
| | - Martin Kruse
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA.
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA.
| | - Byung-Chang Suh
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea.
| |
Collapse
|
49
|
Tang QY, Larry T, Hendra K, Yamamoto E, Bell J, Cui M, Logothetis DE, Boland LM. Mutations in Nature Conferred a High Affinity Phosphatidylinositol 4,5-Bisphosphate-binding Site in Vertebrate Inwardly Rectifying Potassium Channels. J Biol Chem 2015; 290:16517-29. [PMID: 25957411 DOI: 10.1074/jbc.m115.640409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Indexed: 11/06/2022] Open
Abstract
All vertebrate inwardly rectifying potassium (Kir) channels are activated by phosphatidylinositol 4,5-bisphosphate (PIP2) (Logothetis, D. E., Petrou, V. I., Zhang, M., Mahajan, R., Meng, X. Y., Adney, S. K., Cui, M., and Baki, L. (2015) Annu. Rev. Physiol. 77, 81-104; Fürst, O., Mondou, B., and D'Avanzo, N. (2014) Front. Physiol. 4, 404-404). Structural components of a PIP2-binding site are conserved in vertebrate Kir channels but not in distantly related animals such as sponges and sea anemones. To expand our understanding of the structure-function relationships of PIP2 regulation of Kir channels, we studied AqKir, which was cloned from the marine sponge Amphimedon queenslandica, an animal that represents the phylogenetically oldest metazoans. A requirement for PIP2 in the maintenance of AqKir activity was examined in intact oocytes by activation of a co-expressed voltage-sensing phosphatase, application of wortmannin (at micromolar concentrations), and activation of a co-expressed muscarinic acetylcholine receptor. All three mechanisms to reduce the availability of PIP2 resulted in inhibition of AqKir current. However, time-dependent rundown of AqKir currents in inside-out patches could not be re-activated by direct application to the inside membrane surface of water-soluble dioctanoyl PIP2, and the current was incompletely re-activated by the more hydrophobic arachidonyl stearyl PIP2. When we introduced mutations to AqKir to restore two positive charges within the vertebrate PIP2-binding site, both forms of PIP2 strongly re-activated the mutant sponge channels in inside-out patches. Molecular dynamics simulations validate the additional hydrogen bonding potential of the sponge channel mutants. Thus, nature's mutations conferred a high affinity activation of vertebrate Kir channels by PIP2, and this is a more recent evolutionary development than the structures that explain ion channel selectivity and inward rectification.
Collapse
Affiliation(s)
- Qiong-Yao Tang
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, the Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, XuZhou Medical College, Xuzhou, 221004 Jiangsu Province, China
| | - Trevor Larry
- the Department of Biology, University of Richmond, Richmond, Virginia 23173
| | - Kalen Hendra
- the Department of Biology, University of Richmond, Richmond, Virginia 23173
| | - Erica Yamamoto
- the Department of Biology, University of Richmond, Richmond, Virginia 23173
| | - Jessica Bell
- the Department of Chemistry and Biochemistry, University of San Diego, San Diego, California 92110, and
| | - Meng Cui
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Diomedes E Logothetis
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Linda M Boland
- the Department of Biology, University of Richmond, Richmond, Virginia 23173
| |
Collapse
|
50
|
Zhang RS, Wright JD, Pless SA, Nunez JJ, Kim RY, Li JBW, Yang R, Ahern CA, Kurata HT. A Conserved Residue Cluster That Governs Kinetics of ATP-dependent Gating of Kir6.2 Potassium Channels. J Biol Chem 2015; 290:15450-15461. [PMID: 25934393 DOI: 10.1074/jbc.m114.631960] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Indexed: 12/13/2022] Open
Abstract
ATP-sensitive potassium (KATP) channels are heteromultimeric complexes of an inwardly rectifying Kir channel (Kir6.x) and sulfonylurea receptors. Their regulation by intracellular ATP and ADP generates electrical signals in response to changes in cellular metabolism. We investigated channel elements that control the kinetics of ATP-dependent regulation of KATP (Kir6.2 + SUR1) channels using rapid concentration jumps. WT Kir6.2 channels re-open after rapid washout of ATP with a time constant of ∼60 ms. Extending similar kinetic measurements to numerous mutants revealed fairly modest effects on gating kinetics despite significant changes in ATP sensitivity and open probability. However, we identified a pair of highly conserved neighboring amino acids (Trp-68 and Lys-170) that control the rate of channel opening and inhibition in response to ATP. Paradoxically, mutations of Trp-68 or Lys-170 markedly slow the kinetics of channel opening (500 and 700 ms for W68L and K170N, respectively), while increasing channel open probability. Examining the functional effects of these residues using φ value analysis revealed a steep negative slope. This finding implies that these residues play a role in lowering the transition state energy barrier between open and closed channel states. Using unnatural amino acid incorporation, we demonstrate the requirement for a planar amino acid at Kir6.2 position 68 for normal channel gating, which is potentially necessary to localize the ϵ-amine of Lys-170 in the phosphatidylinositol 4,5-bisphosphate-binding site. Overall, our findings identify a discrete pair of highly conserved residues with an essential role for controlling gating kinetics of Kir channels.
Collapse
Affiliation(s)
- Roger S Zhang
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jordan D Wright
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Stephan A Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - John-Jose Nunez
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Robin Y Kim
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jenny B W Li
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Runying Yang
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa 52246
| | - Harley T Kurata
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|