1
|
Carbajal-García A, Reyes-García J, Díaz-Hernández V, Casas-Hernández MF, Flores-Murrieta FJ, Montaño LM. Testosterone Enhances K V Currents and Airway Smooth Muscle Relaxation Induced by ATP and UTP through P2Y 4 Receptors and Adenylyl Cyclase Pathway. Int J Mol Sci 2024; 25:4652. [PMID: 38731872 PMCID: PMC11083821 DOI: 10.3390/ijms25094652] [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: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
Numerous studies suggest the involvement of adenosine-5'-triphosphate (ATP) and similar nucleotides in the pathophysiology of asthma. Androgens, such as testosterone (TES), are proposed to alleviate asthma symptoms in young men. ATP and uridine-5'-triphosphate (UTP) relax the airway smooth muscle (ASM) via purinergic P2Y2 and P2Y4 receptors and K+ channel opening. We previously demonstrated that TES increased the expression of voltage-dependent K+ (KV) channels in ASM. This study investigates how TES may potentiate ASM relaxation induced by ATP and UTP. Tracheal tissues treated with or without TES (control group) from young male guinea pigs were used. In organ baths, tracheas exposed to TES (40 nM for 48 h) showed enhanced ATP- and UTP-evoked relaxation. Tetraethylammonium, a K+ channel blocker, annulled this effect. Patch-clamp experiments in tracheal myocytes showed that TES also increased ATP- and UTP-induced K+ currents, and this effect was abolished with flutamide (an androgen receptor antagonist). KV channels were involved in this phenomenon, which was demonstrated by inhibition with 4-aminopyridine. RB2 (an antagonist of almost all P2Y receptors except for P2Y2), as well as N-ethylmaleimide and SQ 22,536 (inhibitors of G proteins and adenylyl cyclase, respectively), attenuated the enhancement of the K+ currents induced by TES. Immunofluorescence and immunohistochemistry studies revealed that TES did not modify the expression of P2Y4 receptors or COX-1 and COX-2, while we have demonstrated that this androgen augmented the expression of KV1.2 and KV1.5 channels in ASM. Thus, TES leads to the upregulation of P2Y4 signaling and KV channels in guinea pig ASM, enhancing ATP and UTP relaxation responses, which likely limits the severity of bronchospasm in young males.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Verónica Díaz-Hernández
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - María F. Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Francisco Javier Flores-Murrieta
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| |
Collapse
|
2
|
Romero-Martínez BS, Sommer B, Solís-Chagoyán H, Calixto E, Aquino-Gálvez A, Jaimez R, Gomez-Verjan JC, González-Avila G, Flores-Soto E, Montaño LM. Estrogenic Modulation of Ionic Channels, Pumps and Exchangers in Airway Smooth Muscle. Int J Mol Sci 2023; 24:ijms24097879. [PMID: 37175587 PMCID: PMC10178541 DOI: 10.3390/ijms24097879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 05/15/2023] Open
Abstract
To preserve ionic homeostasis (primarily Ca2+, K+, Na+, and Cl-), in the airway smooth muscle (ASM) numerous transporters (channels, exchangers, and pumps) regulate the influx and efflux of these ions. Many of intracellular processes depend on continuous ionic permeation, including exocytosis, contraction, metabolism, transcription, fecundation, proliferation, and apoptosis. These mechanisms are precisely regulated, for instance, through hormonal activity. The lipophilic nature of steroidal hormones allows their free transit into the cell where, in most cases, they occupy their cognate receptor to generate genomic actions. In the sense, estrogens can stimulate development, proliferation, migration, and survival of target cells, including in lung physiology. Non-genomic actions on the other hand do not imply estrogen's intracellular receptor occupation, nor do they initiate transcription and are mostly immediate to the stimulus. Among estrogen's non genomic responses regulation of calcium homeostasis and contraction and relaxation processes play paramount roles in ASM. On the other hand, disruption of calcium homeostasis has been closely associated with some ASM pathological mechanism. Thus, this paper intends to summarize the effects of estrogen on ionic handling proteins in ASM. The considerable diversity, range and power of estrogens regulates ionic homeostasis through genomic and non-genomic mechanisms.
Collapse
Affiliation(s)
- Bianca S Romero-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México 14080, Mexico
| | - Héctor Solís-Chagoyán
- Neurociencia Cognitiva Evolutiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | - Eduardo Calixto
- Departamento de Neurobiología, Dirección de Investigación en Neurociencias, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Ciudad de México 14370, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City 14080, Mexico
| | - Ruth Jaimez
- Laboratorio de Estrógenos y Hemostasis, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Juan C Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), Ciudad de México 10200, Mexico
| | - Georgina González-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", México City 14080, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
3
|
Reyes-García J, Díaz-Hernández V, Carbajal-García A, Casas-Hernández MF, Sommer B, Montaño LM. Theophylline-Induced Relaxation Is Enhanced after Testosterone Treatment via Increased K V1.2 and K V1.5 Protein Expression in Guinea Pig Tracheal Smooth Muscle. Int J Mol Sci 2023; 24:ijms24065884. [PMID: 36982957 PMCID: PMC10059212 DOI: 10.3390/ijms24065884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/16/2023] [Accepted: 02/25/2023] [Indexed: 03/30/2023] Open
Abstract
Theophylline is a drug commonly used to treat asthma due to its anti-inflammatory and bronchodilatory properties. Testosterone (TES) has been suggested to reduce the severity of asthma symptoms. This condition affects boys more than girls in childhood, and this ratio reverses at puberty. We reported that guinea pig tracheal tissue chronic exposure to TES increases the expression of β2-adrenoreceptors and enhances salbutamol-induced K+ currents (IK+). Herein, we investigated whether the upregulation of K+ channels can enhance the relaxation response to methylxanthines, including theophylline. Chronic incubation of guinea pig tracheas with TES (40 nM, 48 h) enhanced the relaxation induced by caffeine, isobutylmethylxanthine, and theophylline, an effect that was abolished by tetraethylammonium. In tracheal myocytes, chronic incubation with TES increased theophylline-induced IK+; flutamide reversed this effect. The increase in IK+ was blocked by 4-aminopyridine by ~82%, whereas iberiotoxin reduced IK+ by ~17%. Immunofluorescence studies showed that chronic TES exposure increased the expression of KV1.2 and KV1.5 in airway smooth muscle (ASM). In conclusion, chronic exposure to TES in guinea pig ASM promotes upregulation of KV1.2 and KV1.5 and enhances theophylline relaxation response. Therefore, gender should be considered when prescribing methylxanthines, as teenage boys and males are likely to respond better than females.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Verónica Díaz-Hernández
- Departamento de Embriología y Genética, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - María F Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City 14080, Mexico
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
4
|
Carbajal-García A, Reyes-García J, Casas-Hernández MF, Flores-Soto E, Díaz-Hernández V, Solís-Chagoyán H, Sommer B, Montaño LM. Testosterone augments β 2 adrenergic receptor genomic transcription increasing salbutamol relaxation in airway smooth muscle. Mol Cell Endocrinol 2020; 510:110801. [PMID: 32278021 DOI: 10.1016/j.mce.2020.110801] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/12/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
Androgens in asthmatic men may be linked to asthma severity, acting via nongenomic and genomic effects. This ailment affects boys more than girls during infancy, and this proportion reverses in puberty. Plasmatic androgen concentration in young men increases at this age and might be related to lower asthma symptoms. Nongenomic actions occur in a brief period and are independent of the androgen receptor (AR), while genomic effects depend on AR, take hours-days and are modified by transcription or protein synthesis inhibitors. Guinea pig tracheas chronic incubation with testosterone (TES, 40 nM, 48 h) potentiates salbutamol-induced relaxation, an effect that was reversed by flutamide, not observed when tissues were pre-incubated with TES-bovine serum albumin (TES-BSA) nor when tissues were preincubated with TES for 15-60 min. In tracheal myocytes, TES chronic incubation increases salbutamol-induced K+ currents (IK+), an effect that was also reversed by flutamide, actinomycin D and cycloheximide and not seen with TES-BSA. The increment in IK+ was blocked by 4-aminopyridine and iberiotoxin, indicating that delayed rectifier K+ and high-conductance Ca2+ activated K+ channels were involved in the TES potentiation effect. Immunofluorescence studies showed that chronic TES augmented the β2 adrenergic receptor (β2-AR) expression in ASM and this finding was corroborated by q-PCR and Western blot assays. β2-AR affinity for salbutamol after TES incubation was increased. In conclusion, chronic exposure to physiological TES concentration of the guinea pig ASM promotes β2-AR upregulation favoring β2 adrenergic responses and probably limiting the severity of the asthmatic exacerbations in teenage boys and men.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - María F Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Verónica Díaz-Hernández
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, CDMX, México
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, CDMX, México
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México.
| |
Collapse
|
5
|
Deng M, Jiang L, Luo X, Tao H, Liang S. Jingzhaotoxin-X, a gating modifier of Kv4.2 and Kv4.3 potassium channels purified from the venom of the Chinese tarantula Chilobrachys jingzhao. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190043. [PMID: 32536941 PMCID: PMC7269146 DOI: 10.1590/1678-9199-jvatitd-2019-0043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background The tarantula Chilobrachys jingzhao is one of the largest venomous spiders in China. In previous studies, we purified and characterized at least eight peptides from C. jingzhao venom. In this report, we describe the purification and characterization of Jingzhaotoxin-X (JZTX-X), which selectively blocks Kv4.2 and Kv4.3 potassium channels. Methods JZTX-X was purified using a combination of cation-exchange HPLC and reverse-phase HPLC. The amino-acid sequence was determined by automated Edman degradation and confirmed by mass spectrometry (MS). Voltage-gated ion channel currents were recorded in HEK293t cells transiently transfected with a variety of ion channel constructs. In addition, the hyperalgesic activity of JZTX-X and the toxin´s effect on motor function were assessed in mice. Results JZTX-X contained 31 amino acids, with six cysteine residues that formed three disulfide bonds within an inhibitory cysteine knot (ICK) topology. In whole-cell voltage-clamp experiments, JZTX-X inhibited Kv4.2 and Kv4.3 potassium channels in a concentration- and voltage-dependent manner, without affecting other ion channels (Kv1.1, 1.2, 1.3, 2.1, delayed rectifier potassium channels, high- and low-voltage-activated Ca2+ channels, and voltage-gated sodium channels Nav1.5 and 1.7). JZTX-X also shifted the voltage-dependent channel activation to more depolarized potentials, whereas extreme depolarization caused reversible toxin binding to Kv4.2 channels. JZTX-X shifted the Kv4.2 and Kv4.3 activities towards a resting state, since at the resting potential the toxin completely inhibited the channels, even in the absence of an applied physical stimulus. Intrathecal or intraplantar injection of JZTX-X caused a long-lasting decrease in the mechanical nociceptive threshold (hyperalgesia) but had no effect on motor function as assessed in the rotarod test. Conclusions JZTX-X selectively suppresses Kv4.2 and Kv4.3 potassium channel activity in a concentration- and voltage-dependent manner and causes long-lasting mechanical hyperalgesia.
Collapse
Affiliation(s)
- Meichun Deng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Liping Jiang
- Department of Parasitology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xuan Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| |
Collapse
|
6
|
Carbajal-García A, Reyes-García J, Montaño LM. Androgen Effects on the Adrenergic System of the Vascular, Airway, and Cardiac Myocytes and Their Relevance in Pathological Processes. Int J Endocrinol 2020; 2020:8849641. [PMID: 33273918 PMCID: PMC7676939 DOI: 10.1155/2020/8849641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Androgen signaling comprises nongenomic and genomic pathways. Nongenomic actions are not related to the binding of the androgen receptor (AR) and occur rapidly. The genomic effects implicate the binding to a cytosolic AR, leading to protein synthesis. Both events are independent of each other. Genomic effects have been associated with different pathologies such as vascular ischemia, hypertension, asthma, and cardiovascular diseases. Catecholamines play a crucial role in regulating vascular smooth muscle (VSM), airway smooth muscle (ASM), and cardiac muscle (CM) function and tone. OBJECTIVE The aim of this review is an updated analysis of the role of androgens in the adrenergic system of vascular, airway, and cardiac myocytes. Body. Testosterone (T) favors vasoconstriction, and its concentration fluctuation during life stages can affect the vascular tone and might contribute to the development of hypertension. In the VSM, T increases α1-adrenergic receptors (α 1-ARs) and decreases adenylyl cyclase expression, favoring high blood pressure and hypertension. Androgens have also been associated with asthma. During puberty, girls are more susceptible to present asthma symptoms than boys because of the increment in the plasmatic concentrations of T in young men. In the ASM, β 2-ARs are responsible for the bronchodilator effect, and T augments the expression of β 2-ARs evoking an increase in the relaxing response to salbutamol. The levels of T are also associated with an increment in atherosclerosis and cardiovascular risk. In the CM, activation of α 1A-ARs and β 2-ARs increases the ionotropic activity, leading to the development of contraction, and T upregulates the expression of both receptors and improves the myocardial performance. CONCLUSIONS Androgens play an essential role in the adrenergic system of vascular, airway, and cardiac myocytes, favoring either a state of health or disease. While the use of androgens as a therapeutic tool for treating asthma symptoms or heart disease is proposed, the vascular system is warmly affected.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico
| |
Collapse
|
7
|
Deng M, Jiang L, Luo X, Tao H, Liang S. Jingzhaotoxin-X, a gating modifier of Kv4.2 and Kv4.3 potassium channels purified from the venom of the Chinese tarantula Chilobrachys jingzhao. J Venom Anim Toxins Incl Trop Dis 2020. [DOI: 10.1590//1678-9199-jvatitd-2019-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | | | | | - Huai Tao
- Hunan University of Chinese Medicine, China
| | | |
Collapse
|
8
|
Simeone KA, Hallgren J, Bockman CS, Aggarwal A, Kansal V, Netzel L, Iyer SH, Matthews SA, Deodhar M, Oldenburg PJ, Abel PW, Simeone TA. Respiratory dysfunction progresses with age in Kcna1-null mice, a model of sudden unexpected death in epilepsy. Epilepsia 2018; 59:345-357. [PMID: 29327348 DOI: 10.1111/epi.13971] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Increased breathing rate, apnea, and respiratory failure are associated with sudden unexpected death in epilepsy (SUDEP). We recently demonstrated the progressive nature of epilepsy and mortality in Kcna1-/- mice, a model of temporal lobe epilepsy and SUDEP. Here we tested the hypothesis that respiratory dysfunction progresses with age in Kcna1-/- mice, thereby increasing risk of respiratory failure and sudden death (SD). METHODS Respiratory parameters were determined in conscious mice at baseline and following increasing doses of methacholine (MCh) using noninvasive airway mechanics (NAM) systems. Kcna1+/+ , Kcna1+/- , and Kcna1-/- littermates were assessed during 3 age ranges when up to ~30%, ~55%, and ~90% of Kcna1-/- mice have succumbed to SUDEP: postnatal day (P) 32-36, P40-46, and P48-56, respectively. Saturated arterial O2 (SaO2 ) was determined with pulse oximetry. Lung and brain tissues were isolated and Kcna1 gene and protein expression were evaluated by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) and Western blot techniques. Airway smooth muscle responsiveness was assessed in isolated trachea exposed to MCh. RESULTS Kcna1-/- mice experienced an increase in basal respiratory drive, chronic oxygen desaturation, frequent apnea-hypopnea (A-H), an atypical breathing sequence of A-H-tachypnea-A-H, increased tidal volume, and hyperventilation induced by MCh. The MCh-provoked hyperventilation was dramatically attenuated with age. Of interest, only Kcna1-/- mice developed seizures following exposure to MCh. Seizures were provoked by lower concentrations of MCh as Kcna1-/- mice approached SD. MCh-induced seizures experienced by a subset of younger Kcna1-/- mice triggered death. Respiratory parameters of these younger Kcna1-/- mice resembled older near-SD Kcna1-/- mice. Kcna1 gene and protein were not expressed in Kcna1+/+ and Kcna1+/- lungs, and MCh-mediated airway smooth muscle contractions exhibited similar half-maximal effective concentration( EC50 ) in isolated Kcna1+/+ and Kcna1-/- trachea. SIGNIFICANCE The Kcna1-/- model of SUDEP exhibits progressive respiratory dysfunction, which suggests a potential increased susceptibility for respiratory failure during severe seizures that may result in sudden death.
Collapse
Affiliation(s)
- Kristina A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Jodi Hallgren
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Charles S Bockman
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Ankita Aggarwal
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Vikash Kansal
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Lauren Netzel
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Shruthi H Iyer
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Stephanie A Matthews
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Malavika Deodhar
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Peter J Oldenburg
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter W Abel
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Timothy A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
9
|
Xiao GS, Zhang YH, Wu W, Sun HY, Wang Y, Li GR. Genistein and tyrphostin AG556 decrease ultra-rapidly activating delayed rectifier K + current of human atria by inhibiting EGF receptor tyrosine kinase. Br J Pharmacol 2017; 174:454-467. [PMID: 28072464 DOI: 10.1111/bph.13710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE The ultra-rapidly activating delayed rectifier K+ current IKur (encoded by Kv 1.5 or KCNA5) plays an important role in human atrial repolarization. The present study investigates the regulation of this current by protein tyrosine kinases (PTKs). EXPERIMENTAL APPROACH Whole-cell patch voltage clamp technique and immunoprecipitation and Western blotting analysis were used to investigate whether the PTK inhibitors genistein, tyrphostin AG556 (AG556) and PP2 regulate human atrial IKur and hKv1.5 channels stably expressed in HEK 293 cells. KEY RESULTS Human atrial IKur was decreased by genistein (a broad-spectrum PTK inhibitor) and AG556 (a highly selective EGFR TK inhibitor) in a concentration-dependent manner. Inhibition of IKur induced by 30 μM genistein or 10 μM AG556 was significantly reversed by 1 mM orthovanadate (a protein tyrosine phosphatase inhibitor). Similar results were observed in HEK 293 cells stably expressing hKv 1.5 channels. On the other hand, the Src family kinase inhibitor PP2 (1 μM) slightly enhanced IKur and hKv 1.5 current, and the current increase was also reversed by orthovanadate. Immunoprecipitation and Western blotting analysis showed that genistein, AG556, and PP2 decreased tyrosine phosphorylation of hKv 1.5 channels and that the decrease was countered by orthovanadate. CONCLUSION AND IMPLICATIONS The PTK inhibitors genistein and AG556 decrease human atrial IKur and cloned hKv 1.5 channels by inhibiting EGFR TK, whereas the Src kinase inhibitor PP2 increases IKur and hKv 1.5 current. These results imply that EGFR TK and the soluble Src kinases may have opposite effects on human atrial IKur .
Collapse
Affiliation(s)
- Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yan-Hui Zhang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wei Wu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hai-Ying Sun
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
10
|
Gutterman DD, Chabowski DS, Kadlec AO, Durand MJ, Freed JK, Ait-Aissa K, Beyer AM. The Human Microcirculation: Regulation of Flow and Beyond. Circ Res 2016; 118:157-72. [PMID: 26837746 DOI: 10.1161/circresaha.115.305364] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The microcirculation is responsible for orchestrating adjustments in vascular tone to match local tissue perfusion with oxygen demand. Beyond this metabolic dilation, the microvasculature plays a critical role in modulating vascular tone by endothelial release of an unusually diverse family of compounds including nitric oxide, other reactive oxygen species, and arachidonic acid metabolites. Animal models have provided excellent insight into mechanisms of vasoregulation in health and disease. However, there are unique aspects of the human microcirculation that serve as the focus of this review. The concept is put forth that vasculoparenchymal communication is multimodal, with vascular release of nitric oxide eliciting dilation and preserving normal parenchymal function by inhibiting inflammation and proliferation. Likewise, in disease or stress, endothelial release of reactive oxygen species mediates both dilation and parenchymal inflammation leading to cellular dysfunction, thrombosis, and fibrosis. Some pathways responsible for this stress-induced shift in mediator of vasodilation are proposed. This paradigm may help explain why microvascular dysfunction is such a powerful predictor of cardiovascular events and help identify new approaches to treatment and prevention.
Collapse
Affiliation(s)
- David D Gutterman
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee.
| | - Dawid S Chabowski
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Andrew O Kadlec
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Matthew J Durand
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Julie K Freed
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Karima Ait-Aissa
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Andreas M Beyer
- From the Cardiovascular Center (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A., M.J.D.), Departments of Medicine (A.M.B., A.O.K., D.D.G., D.S.C., J.K.F., K.A.-A.), Pharmacology and Toxicology (D.S.C., J.K.F.), Physiology (A.M.B., A.O.K.), Physical Medicine and Rehabilitation (M.J.D.), and Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
11
|
Andersen MN, Skibsbye L, Tang C, Petersen F, MacAulay N, Rasmussen HB, Jespersen T. PKC and AMPK regulation of Kv1.5 potassium channels. Channels (Austin) 2016; 9:121-8. [PMID: 26043299 DOI: 10.1080/19336950.2015.1036205] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The voltage-gated Kv1.5 potassium channel, conducting the ultra-rapid rectifier K(+) current (IKur), is regulated through several pathways. Here we investigate if Kv1.5 surface expression is controlled by the 2 kinases PKC and AMPK, using Xenopus oocytes, MDCK cells and atrial derived HL-1 cells. By confocal microscopy combined with electrophysiology we demonstrate that PKC activation reduces Kv1.5 current, through a decrease in membrane expressed channels. AMPK activation was found to decrease the membrane expression in MDCK cells, but not in HL-1 cells and was furthermore shown to be dependent on co-expression of Nedd4-2 in Xenopus oocytes. These results indicate that Kv1.5 channels are regulated by both kinases, although through different molecular mechanisms in different cell systems.
Collapse
Affiliation(s)
- Martin Nybo Andersen
- a Danish National Research Foundation Center for Cardiac Arrhythmia; Dept. of Biomedical Sciences ; Faculty of Health and Medical Sciences; University of Copenhagen ; Copenhagen , Denmark
| | | | | | | | | | | | | |
Collapse
|
12
|
Hu Z, Ma A, Zhang Y, Xi Y, Fan L, Wang T, Zhang T. Voltage-gated potassium+ channel expression in coronary artery smooth muscle cells of SHR and WKY. Cell Biochem Biophys 2015; 70:1725-31. [PMID: 25030407 DOI: 10.1007/s12013-014-0120-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study aims to compare the expression of genes and the molecular characteristic of voltage-gated K(+) channels, which make great effort in maintaining and controlling smooth muscle contraction, cellular membrane potential, and intracellular calcium ion currents in artery smooth muscle cells of SHR and WKY. Expression of potassium ions family in coronary artery was detected through reverse transcription polymerase chain reaction quantitatively. Significant levels of voltage-gated K(+) channels α1.2, α1.5, and β1.1 expression were all proved to be significantly higher in smooth muscles of SHR than WKY. Whole-cell voltage-gated K(+) channel currents were larger in SHR artery smooth muscles than the ones of WKY. Moreover, the voltage dependence of voltage-gated potassium channel activation was more negative in artery smooth muscle of SHR than that of WKY, while voltage dependence of availability was not different. The above diversity of voltage-gated potassium channel detected in gene expression and electrical character in coronary artery smooth muscle of SHR than that of WKY might be an underling mechanism associated with the membrane potential depolarization in artery smooth muscle of SHR.
Collapse
Affiliation(s)
- Zhi Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Pan Y, Levin EJ, Quick M, Zhou M. Potentiation of the Kv1 family K(+) channel by cortisone analogues. ACS Chem Biol 2012; 7:1641-6. [PMID: 22803826 DOI: 10.1021/cb300233y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Kv1 family voltage-dependent K(+) channels are essential for termination of action potentials in neurons and myocytes. These channels form a stable complex with their beta subunits (Kvβ), some of which inhibit channel activity. Cortisone potentiates Kv1 channel by binding to Kvβ and promoting its dissociation from the channel, but its half-maximum effective concentration is ∼46 μM. To identify corticosteroids that are more efficient than cortisone, we examined 25 cortisone analogues and found that fluticasone propionate potentiates channel current with a half-maximum effective concentration (EC(50)) of 37 ± 1.1 nM. Further studies showed that fluticasone propionate potentiates channel current by inducing dissociation of Kvβ, and docking of fluticasone propionate into the cortisone binding site reveals potential interactions that enhance the EC(50) value. Thus, fluticasone propionate provides a starting point for rational design of more efficient small-molecule compounds that increase Kv1 activity and affect the integrity of the Kv1-Kvβ complex.
Collapse
Affiliation(s)
| | | | - Matthias Quick
- Division
of Molecular Therapeutics, New York State Psychiatric Institute, 1051 Riverside
Drive, New York, New York 10032, United States
| | | |
Collapse
|
14
|
Mia S, Munoz C, Pakladok T, Siraskar G, Voelkl J, Alesutan I, Lang F. Downregulation of Kv1.5 K channels by the AMP-activated protein kinase. Cell Physiol Biochem 2012; 30:1039-50. [PMID: 23221389 DOI: 10.1159/000341480] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2012] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The voltage gated K(+) channel Kv1.5 participates in the repolarization of a wide variety of cell types. Kv1.5 is downregulated during hypoxia, which is known to stimulate the energy-sensing AMP-activated serine/threonine protein kinase (AMPK). AMPK is a powerful regulator of nutrient transport and metabolism. Moreover, AMPK is known to downregulate several ion channels, an effect at least in part due to stimulation of the ubiquitin ligase Nedd4- 2. The present study explored whether AMPK regulates Kv1.5. METHODS cRNA encoding Kv1.5 was injected into Xenopus oocytes with and without additional injection of wild-type AMPK (α1 β 1γ1), of constitutively active (γR70Q)AMPK (α1 β 1γ1(R70Q)), of inactive mutant (αK45R)AMPK (α1(K45R)β1γ1), or of Nedd4-2. Kv1.5 activity was determined by two-electrode voltage-clamp. Moreover, Kv1.5 protein abundance in the cell membrane was determined by chemiluminescence and immunostaining with subsequent confocal microscopy. RESULTS Coexpression of wild-type AMPK(WT) and constitutively active AMPK(γR70Q), but not of inactive AMPK(αK45R) significantly reduced Kv1.5-mediated currents. Coexpression of constitutively active AMPKγR70Q further reduced Kv1.5 K(+) channel protein abundance in the cell membrane. Co-expression of Nedd4-2 similarly downregulated Kv1.5-mediated currents. CONCLUSION AMPK is a potent regulator of Kv1.5. AMPK inhibits Kv1.5 presumably in part by activation of Nedd4- 2 with subsequent clearance of channel protein from the cell membrane.
Collapse
Affiliation(s)
- Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Munoz C, Tóvolli RH, Sopjani M, Alesutan I, Lam RS, Seebohm G, Föller M, Lang F. Activation of voltage gated K⁺ channel Kv1.5 by β-catenin. Biochem Biophys Res Commun 2011; 417:692-6. [PMID: 22166221 DOI: 10.1016/j.bbrc.2011.11.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 11/30/2011] [Indexed: 01/16/2023]
Abstract
Voltage-gated Kv1.5 channels are expressed in a wide variety of tissues including cardiac myocytes, smooth muscle and tumor cells. Kv1.5 channel activity is modified by N-cadherin, which in turn binds the multifunctional oncogenic protein β-catenin. The present experiments explored the effect of β-catenin on Kv1.5 channel activity. To this end, Kv1.5 was expressed in Xenopus oocytes with or without β-catenin and the voltage-gated Kv current determined by dual electrode voltage clamp. As a result, expression of β-catenin significantly increased the voltage-gated Kv current at positive potentials. The stimulating effect of β-catenin on Kv1.5 was not dependent on the stimulation of transcription since it was observed even in the presence of the transcription inhibitor actinomycin D. Specific antibody binding to surface Kv1.5 in Xenopus oocytes revealed that β-catenin enhances the membrane abundance of Kv1.5. Further experiments with brefeldin A showed that β-catenin fosters the insertion of Kv1.5 into rather than delaying the retrieval from the plasma membrane. According to electrophysiological recordings with mutant β-catenin, the effect on Kv1.5 requires the same protein domains that are required for association of β-catenin with cadherin. The experiments disclose a completely novel function of β-catenin, i.e. the regulation of Kv1.5 channel activity.
Collapse
Affiliation(s)
- Carlos Munoz
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Stirling L, Williams MR, Morielli AD. Dual roles for RHOA/RHO-kinase in the regulated trafficking of a voltage-sensitive potassium channel. Mol Biol Cell 2009; 20:2991-3002. [PMID: 19403695 DOI: 10.1091/mbc.e08-10-1074] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Kv1.2 is a member of the Shaker family of voltage-sensitive potassium channels and contributes to regulation of membrane excitability. The electrophysiological activity of Kv1.2 undergoes tyrosine kinase-dependent suppression in a process involving RhoA. We report that RhoA elicits suppression of Kv1.2 ionic current by modulating channel endocytosis. This occurs through two distinct pathways, one clathrin-dependent and the other cholesterol-dependent. Activation of Rho kinase (ROCK) via the lysophosphatidic acid (LPA) receptor elicits clathrin-dependent Kv1.2 endocytosis and consequent attenuation of its ionic current. LPA-induced channel endocytosis is blocked by the ROCK inhibitor Y27632 or by clathrin RNA interference. In contrast, steady-state endocytosis of Kv1.2 in unstimulated cells is cholesterol dependent. Inhibition of basal ROCK signaling with Y27632 increased surface Kv1.2, an effect that persists in the presence of clathrin small interfering RNA and that is not additive to the increase in surface channel levels elicited by the cholesterol sequestering drug filipin. Temperature block experiments show that ROCK affects cholesterol-dependent trafficking by modulating the recycling of endocytosed channel back to the plasma membrane. Both receptor-stimulated and steady-state Kv1.2 trafficking modulated by RhoA/ROCK required the activation of dynamin as well as the ROCK effector Lim-kinase, indicating a key role for actin remodeling in RhoA-dependent Kv1.2 regulation.
Collapse
Affiliation(s)
- Lee Stirling
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | |
Collapse
|
17
|
Cheng D, Xu Y, Liu X, Zhao L, Xiong S, Zhang Z. The effects of protein kinase C (PKC) on the tension of normal and passively sensitized human airway smooth muscle and the activity of voltage-dependent delayed rectifier potassium channel (Kv). ACTA ACUST UNITED AC 2008; 27:153-6. [PMID: 17497283 DOI: 10.1007/s11596-007-0211-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Indexed: 11/26/2022]
Abstract
The effects of protein kinase C (PKC) on the tension and the activity of voltage-dependent delayed rectifier potassium channel (K(y)) were examined in normal and passively sensitized human airway smooth muscle (HASM), by measuring tones and whole-cell patch clamp techniques, and the K(v) activities and membrane potential (E (m)) were also detected. The results showed that phorbol 12-myristate 13-acetate (PMA), a PKC activator, caused a concentration-dependent constriction in normal HASM rings. The constriction of the passively sensitized muscle in asthma serum group was significantly higher than that of the normal group (P<0.05), and the constrictions of both groups were completely abolished by PKC inhibitor Ro31-8220 and calcium channel inhibitor nifedipine. K(v) activities of HASM cells were significantly inhibited by PMA, and the E (m) became more positive, as compared with the DMSO (a PMA menstruum)-treated group (P<0.01). This effect could be blocked by Ro31-8220 (P<0.01). It was concluded that activation of PKC could increase the tones of HASM, which might be related to the reduced K(v) activity. In passively sensitized HASM rings, this effect was more notable.
Collapse
Affiliation(s)
- Dongjun Cheng
- Department of Respiratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
18
|
Yuan C, Liao Z, Zeng X, Dai L, Kuang F, Liang S. Jingzhaotoxin-XII, a gating modifier specific for Kv4.1 channels. Toxicon 2007; 50:646-52. [PMID: 17631373 DOI: 10.1016/j.toxicon.2007.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 05/15/2007] [Accepted: 05/29/2007] [Indexed: 01/17/2023]
Abstract
Jingzhaotoxin-XII (JZTX-XII), a 29-residue polypeptide, was purified from the venom of the Chinese tarantula Chilobrachys jingzhao. Electrophysiological recordings carried out in Xenopus laevis oocytes showed that JZTX-XII is specific for Kv4.1 channels, with the IC50 value of 0.363 microM. It interacts with the channels by modifying the gating behavior. JZTX-XII shares 80% sequence identity with phrixotoxin1, a potent inhibitor for Kv4.2 and Kv4.3 channels. Structure analysis indicates that the difference of the charge distribution in the interactive surface perhaps influences the specific pharmacology of the toxins. JZTX-XII should be a valuable tool for the investigation of the Kv4.1 channels.
Collapse
Affiliation(s)
- Chunhua Yuan
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Life Science College, Hunan Normal University, Changsha 410081, PR China
| | | | | | | | | | | |
Collapse
|
19
|
Koutsouki E, Lam RS, Seebohm G, Ureche ON, Ureche L, Baltaev R, Lang F. Modulation of human Kv1.5 channel kinetics by N-cadherin. Biochem Biophys Res Commun 2007; 363:18-23. [PMID: 17868645 DOI: 10.1016/j.bbrc.2007.07.181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 07/27/2007] [Indexed: 11/25/2022]
Abstract
Kv1.5 is expressed in multiple tissues including heart, brain, macrophages, as well as vascular, airway, and intestinal smooth muscle cells. Kv1.5 currents contribute to cardiac repolarization. In cardiac myocytes Kv1.5 colocalizes with N-cadherin. As Kv1.5 expression increases following establishment of cell-cell contacts and N-cadherin influences the activity of other ion channels, we explored whether N-cadherin participates in the regulation of Kv1.5 activity. To this end, we expressed Kv1.5 in Xenopus oocytes with or without additional expression of N-cadherin. Coexpression of N-cadherin was followed by a approximately 2- to 3-fold increase of Kv1.5 induced current. The effect of N-cadherin was not paralleled by significant alterations of Kv1.5 channel abundance within the oocyte cell membrane but resulted primarily from accelerated recovery from inactivation. In conclusion, N-cadherin modifies Kv1.5 channel activity and is thus a novel candidate signaling molecule participating in the regulation of a variety of functions including cardiac action potential and vascular tone.
Collapse
Affiliation(s)
- Evgenia Koutsouki
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
20
|
Graça C, Baggio CH, Freitas CS, Rattmann YD, de Souza LM, Cipriani TR, Sassaki GL, Rieck L, Pontarolo R, da Silva-Santos JE, Marques MCA. In vivo assessment of safety and mechanisms underlying in vitro relaxation induced by Mikania laevigata Schultz Bip. ex Baker in the rat trachea. JOURNAL OF ETHNOPHARMACOLOGY 2007; 112:430-9. [PMID: 17573215 DOI: 10.1016/j.jep.2007.03.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2006] [Revised: 03/17/2007] [Accepted: 03/24/2007] [Indexed: 05/15/2023]
Abstract
Mikania laevigata, popularly known in Brazil as "guaco", is largely used in folk medicine against respiratory diseases. However, neither the assessment of the toxicity of "guaco" syrup (GS, used by humans) nor its efficacy or mechanisms of action has been properly investigated. Using in vitro procedures, we showed that the hydroalcoholic extract (HE) from Mikania laevigata induces a concentration-dependent relaxation of rat trachea which does not depend on epithelium-derived substances but involves changes in the cellular mobilization of calcium, perhaps due to a direct effect on membrane potassium channels. In addition, we assessed both oral and intraperitoneal acute toxicity, as well as the oral subchronic and chronic toxicity of GS containing controlled amounts of coumarin, the main biological marker of Mikania laevigata preparations used in humans. The calculated LD(50) of GS after intraperitoneal administration was 0.904 g/kg in mice (both sexes) and 0.967 and 0.548 g/kg in male and female rats, respectively. However, the LD(50) values of GS by the oral route were calculated to be up to 10 g/kg, in both male and female mice and rats. Repeated dose 28- or 90-day oral treatment with GS (75, 150 and 300 mg/kg) did not produce any disturbances in the hematological or biochemical parameters of either male or female rats, nor did it provide evidence of toxicity in the hepatic, renal or pancreatic systems. Besides the mechanistic findings, our results provide evidence of the safety of Mikania laevigata in rodents, even after subchronic and chronic administration, at least in relation to the evaluated parameters.
Collapse
Affiliation(s)
- Carlos Graça
- Department of Pharmacy, Universidade Federal do Pará, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Park SA, Lee YC, Ma TZ, Park JA, Han MK, Lee HH, Kim HG, Kwak YG. hKv1.5 channels play a pivotal role in the functions of human alveolar macrophages. Biochem Biophys Res Commun 2006; 346:567-71. [PMID: 16765315 DOI: 10.1016/j.bbrc.2006.05.149] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 05/24/2006] [Indexed: 11/25/2022]
Abstract
We examined the pharmacological properties, the molecular identity, and the functional roles of hKv1.5 channel in human alveolar macrophage. Some of outward K(+) current was inhibited by 4-aminopyridine and antisense oligodeoxynucleotides against hKv1.5 mRNA. Consistently, the protein and mRNA expressions of hKv1.5 channel were detected. Furthermore, the phagocytosis and migration of human alveolar macrophages were significantly suppressed when the protein expression of hKv1.5 channel was lowered by the antisense hKv1.5 oligodeoxynucleotides. These results suggest that hKv1.5 channel is expressed in human alveolar macrophages and it plays a role in phagocytosis and migration of the human alveolar macrophage.
Collapse
Affiliation(s)
- Seon-Ah Park
- Department of Pharmacology, Chonbuk National University Medical School, Jeonju, Jeonbuk, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Plante I, Fournier D, Ricard G, Drolet B, O'Hara G, Champagne J, Mathieu P, Baillot R, Daleau P. Electrophysiological characterization of three non-synonymous single nucleotide polymorphisms (R87Q, A251T, and P307S) found in hKv1.5. Pflugers Arch 2006; 452:316-23. [PMID: 16411137 DOI: 10.1007/s00424-005-0031-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 12/01/2005] [Indexed: 10/25/2022]
Abstract
Non-synonymous single nucleotide polymorphisms (SNPs) in the KCNA5/hKv1.5 gene, which encodes for a voltage-gated K+ channel responsible for the I (Kur) current in the human atria, have been recently reported. To gain further knowledge on potential influence of hKv1.5 SNPs, we searched for their presence in a specific population of 96 French-Canadians and characterized electrophysiological properties of the variants in two cell lines. The presumed promoter (-83 bp) and coding regions were sequenced. We found three heterozygous SNPs: R87Q, A251T, and P307S. Functional analysis of SNPs transfected in Chinese hamster ovary (CHO) cells showed that both R87Q and P307S diminished the inactivation amplitude (e.g., at +60 mV, amplitudes were 89+/-26, 23+/-4, and 22+/-7 pA/pF for the wild type, R87Q and P307S, respectively; n=8, 6, and 8, respectively). Inactivation was slowed with these variants (e.g., tau (fast) at +50 mV were 270+/-48, 490+/-66, and 340+/-45 ms for the wild type, R87Q, and P307S, respectively) while R87Q additionally accelerated the rate of hKv1.5 channel opening. A dominant-negative effect was observed for R87Q but not for P307S. SNPs properties were not reproduced when expressed in the HEK293 cell line, suggesting that the regulatory beta-subunit present in CHO cells (and the human heart) is essential for the SNPs effects that we have observed.
Collapse
Affiliation(s)
- Isabelle Plante
- Quebec Heart Institute, Laval Hospital Research Centre, and Faculty of Pharmacy, Laval University, Sainte-Foy, Quebec, G1V 4G5, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Platoshyn O, Brevnova EE, Burg ED, Yu Y, Remillard CV, Yuan JXJ. Acute hypoxia selectively inhibits KCNA5 channels in pulmonary artery smooth muscle cells. Am J Physiol Cell Physiol 2005; 290:C907-16. [PMID: 16236819 PMCID: PMC1363730 DOI: 10.1152/ajpcell.00028.2005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acute hypoxia causes pulmonary vasoconstriction in part by inhibiting voltage-gated K(+) (Kv) channel activity in pulmonary artery smooth muscle cells (PASMC). The hypoxia-mediated decrease in Kv currents [I(K(V))] is selective to PASMC; hypoxia has little effect on I(K(V)) in mesenteric artery smooth muscle cells (MASMC). Functional Kv channels are homo- and/or heterotetramers of pore-forming alpha-subunits and regulatory beta-subunits. KCNA5 is a Kv channel alpha-subunit that forms functional Kv channels in PASMC and regulates resting membrane potential. We have shown that acute hypoxia selectively inhibits I(K(V)) through KCNA5 channels in PASMC. Overexpression of the human KCNA5 gene increased I(K(V)) and caused membrane hyperpolarization in HEK-293, COS-7, and rat MASMC and PASMC. Acute hypoxia did not affect I(K(V)) in KCNA5-transfected HEK-293 and COS-7 cells. However, overexpression of KCNA5 in PASMC conferred its sensitivity to hypoxia. Reduction of Po(2) from 145 to 35 mmHg reduced I(K(V)) by approximately 40% in rat PASMC transfected with human KCNA5 but had no effect on I(K(V)) in KCNA5-transfected rat MASMC (or HEK and COS cells). These results indicate that KCNA5 is an important Kv channel that regulates resting membrane potential and that acute hypoxia selectively reduces KCNA5 channel activity in PASMC relative to MASMC and other cell types. Because Kv channels (including KCNA5) are ubiquitously expressed in PASMC and MASMC, the observation from this study indicates that a hypoxia-sensitive mechanism essential for inhibiting KCNA5 channel activity is exclusively present in PASMC. The divergent effect of hypoxia on I(K(V)) in PASMC and MASMC also may be due to different expression levels of KCNA5 channels.
Collapse
Affiliation(s)
| | | | | | | | | | - Jason X.-J. Yuan
- Address correspondence to: Jason X.-J. Yuan, M.D., Ph.D., Department of Medicine, University of California, San Diego, 9200 Gilman Drive, La Jolla, CA 92093-0725, Tel: (858) 822-6534, Fax: (858) 822-6531, E-mail:
| |
Collapse
|
24
|
Hao K, Niu T, Xu X, Fang Z, Xu X. Single-nucleotide polymorphisms of the KCNS3 gene are significantly associated with airway hyperresponsiveness. Hum Genet 2005; 116:378-83. [PMID: 15714333 DOI: 10.1007/s00439-005-1256-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2004] [Accepted: 12/23/2004] [Indexed: 10/25/2022]
Abstract
Airway hyperresponsiveness (AHR) is one of the major clinical symptoms and intermediate phenotypes of asthma. A recent genome-wide search for asthma quantitative trait loci has revealed a significant linkage signal between a p-terminal region of chromosome 2 and AHR. Thus, the gene encoding the potassium voltage-gated channel delayed-rectifier protein S3 (KCNS3) in this region is considered a positional candidate for asthma. We have evaluated a total of 12 single-nucleotide polymorphisms (SNPs) of the KCNS3 gene in a validation panel of 48 lymphoblastoid cell line DNA samples of Chinese origin. Three SNPs were found to be polymorphic and were tested. Two independent sets (an initial screening set and a replication set) of cases and controls from the original linkage study sample were collected. In the initial screening set, two SNPs (rs1031771 and rs1031772) showed suggestive association and were further confirmed by the replication set. In combined single-SNP analysis, the rs1031771 G allele (odds ratio=1.42, P=0.006) and rs1031772 T allele (odds ratio=1.40, P=0.018) were associated with a significantly higher risk of AHR. Haplotype analysis also detected significant association (P=0.006). Our findings suggest that SNPs located at the 3' downstream region of KCNS3 have a significant role in the etiology of AHR.
Collapse
Affiliation(s)
- Ke Hao
- Program for Population Genetics, Harvard School of Public Health, 665 Huntington Avenue FXB-101, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
25
|
Kwan DCH, Eduljee C, Lee L, Zhang S, Fedida D, Kehl SJ. The external K+ concentration and mutations in the outer pore mouth affect the inhibition of kv1.5 current by Ni2+. Biophys J 2004; 86:2238-50. [PMID: 15041663 PMCID: PMC1304074 DOI: 10.1016/s0006-3495(04)74282-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
By examining the consequences both of changes of [K+]o and of point mutations in the outer pore mouth, our goal was to determine if the mechanism of the block of Kv1.5 ionic currents by external Ni2+ is similar to that for proton block. Ni2+ block is inhibited by increasing [K+]o, by mutating a histidine residue in the pore turret (H463Q) or by mutating a residue near the pore mouth (R487V) that is the homolog of Shaker T449. Aside from a slight rightward shift of the Q-V curve, Ni2+ had no effect on gating currents. We propose that, as with Ho+, Ni2+ binding to H463 facilitates an outer pore inactivation process that is antagonized by Ko+ and that requires R487. However, whereas Ho+ substantially accelerates inactivation of residual currents, Ni2+ is much less potent, indicating incomplete overlap of the profiles of these two metal ions. Analyses with Co2+ and Mn2+, together with previous results, indicate that for the first-row transition metals the rank order for the inhibition of Kv1.5 in 0 mM Ko+ is Zn2+ (KD approximately 0.07 mM) > or = Ni2+) (KD approximately 0.15 mM) > Co2+ (KD approximately 1.4 mM) > Mn2+ (KD > 10 mM).
Collapse
Affiliation(s)
- Daniel C H Kwan
- Department of Physiology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | | | | | | | |
Collapse
|
26
|
Grunnet M, Rasmussen HB, Hay-Schmidt A, Rosenstierne M, Klaerke DA, Olesen SP, Jespersen T. KCNE4 is an inhibitory subunit to Kv1.1 and Kv1.3 potassium channels. Biophys J 2003; 85:1525-37. [PMID: 12944270 PMCID: PMC1303329 DOI: 10.1016/s0006-3495(03)74585-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Kv1 potassium channels are widely distributed in mammalian tissues and are involved in a variety of functions from controlling the firing rate of neurons to maturation of T-lymphocytes. Here we show that the newly described KCNE4 beta-subunit has a drastic inhibitory effect on currents generated by Kv1.1 and Kv1.3 potassium channels. The inhibition is found on channels expressed heterologously in both Xenopus oocytes and mammalian HEK293 cells. mKCNE4 does not inhibit Kv1.2, Kv1.4, Kv1.5, or Kv4.3 homomeric complexes, but it does significantly reduce current through Kv1.1/Kv1.2 and Kv1.2/Kv1.3 heteromeric complexes. Confocal microscopy and Western blotting reveal that Kv1.1 is present at the cell surface together with KCNE4. Real-time RT-PCR shows a relatively high presence of mKCNE4 mRNA in several organs, including uterus, kidney, lung, intestine, and in embryo, whereas a much lower mRNA level is detected in the heart and in five different parts of the brain. Having the broad distribution of Kv1 channels in mind, the demonstrated inhibitory property of KCNE4-subunits could locally and/or transiently have a dramatic influence on cellular excitability and on setting resting membrane potentials.
Collapse
Affiliation(s)
- Morten Grunnet
- Department of Medical Physiology, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | | | | | | | | | | | | |
Collapse
|
27
|
Hirota K, Hashiba E, Yoshioka H, Kabara S, Matsuki A. Effects of three different L-type Ca2+ entry blockers on airway constriction induced by muscarinic receptor stimulation. Br J Anaesth 2003; 90:671-5. [PMID: 12697597 DOI: 10.1093/bja/aeg118] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The crucial role of L-type Ca(2+) channels in airway smooth muscle contraction suggests that these channels could be an important therapeutic target. There are three separate drug binding sites on this channel: those for dihydropyridines, benzothiazepines and phenyl alkylamines. In this study, we examined the effects of the dihydropyridines nifedipine and nicardipine, the benzothiazepine diltiazem, and the phenylalkylamine verapamil on airway constriction. METHODS Tension of guinea-pig tracheal strips was measured isometrically in vitro with a force displacement transducer. Strips were precontracted with carbachol 10(-7) M with or without 4-aminopyridine 10(-3) M, a voltage-sensitive K(+ )channel blocker. Then, nifedipine 10(-8)-10(-4) M, diltiazem 10(-8)-3 x 10(-4) M or verapamil 10(-8)-3 x 10(-4) M was added cumulatively to the organ bath (n=6 each). The bronchial cross-sectional area of pentobarbital-anaesthetized dogs was assessed using a bronchoscopy method. Bronchoconstriction was elicited with methacholine 0.5 micro g kg(-1) plus 5 micro g kg(-1) min(-1), and then nicardipine 0-1000 micro g kg(-1), diltiazem 0-3000 micro g kg(-1) or verapamil 0-3000 micro g kg(-1) were given i.v. (n=7 each). RESULTS In the in vitro experiments, nifedipine and diltiazem fully reversed carbachol-mediated tracheal contraction with logIC(50) values of 4.76 (SEM 0.22) (mean 17.5 micro M) and 4.60 (0.33) (mean 24.8 micro M), respectively. Although verapamil 10(-6)-10(-4) M reversed the contraction by 87.2%, strip tension re-increased by 18.1% following maximal relaxation with verapamil 3 x 10(-4 )M. This re-increase was almost fully abolished by pretreatment with 4-aminopyridine. In the in vivo experiments, nicardipine and diltiazem dose-dependently reversed methacholine-induced bronchoconstriction, with logID(50) values of 3.22 (0.05) (mean 0.60 mg kg(-1)) and 1.85 (0.32) (mean 14.0 mg kg(-1)), respectively. Verapamil worsened methacholine-induced bronchoconstriction. CONCLUSIONS Although supraclinical doses of dihydropyridines and benzothiazepines can produce airway relaxant effects, these agents are unlikely to be used in the treatment of bronchoconstriction. In addition, verapamil may aggravate airway constriction.
Collapse
Affiliation(s)
- K Hirota
- Department of Anesthesiology, University of Hirosaki, School of Medicine, Japan.
| | | | | | | | | |
Collapse
|
28
|
Liu X, Xu Y, Zhang Z, Ni W. K+ channels and their effects on membrane potential in rat bronchial smooth muscle cells. Curr Med Sci 2003; 23:141-4, 150. [PMID: 12973931 DOI: 10.1007/bf02859938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2002] [Indexed: 11/29/2022]
Abstract
In order to investigate the K+ channels and their effects on resting membrane potential (Em) and excitability in rat bronchial smooth muscle cells (BSMCs), the components of outward K+ channel currents and the effects of K+ channels on Em and tension in rat bronchial smooth muscle were observed by using standard whole-cell recording of patch clamp and isometric tension recording techniques. The results showed that under resting conditions, total outward K+ channel currents in freshly isolated BSMCs were unaffected by ATP-sensitive K+ channel blocker. There were two types of K+ currents: voltage-dependent delayed rectifier K+ channel (Kv) and large conductance calcium-activated K+ channel (BKCa) currents. 1 mmol/L 4-aminopyridine (4-AP, an inhibitor of Kv) caused a significant depolarization (from -8.7 +/- 5.9 mV to -25.4 +/- 3.1 mV, n = 18, P < 0.001). In contrast, 1 mmol/L tetraethylammonium (TEA, an inhibitor of BKca) had no significant effect on Em (from -37.6 +/- 4.8 mV to -36.8 +/- 4.1 mV, n = 12, P > 0.05). 4-AP caused a concentration-dependent contraction in resting bronchial strips. TEA had no effect on resting tension, but application of 5 mmol/L TEA resulted in a left shift with bigger pD2 (the negative logarithm of the drug concentration causing 50% of maximal effect) (from 6.27 +/- 0.38 to 6.89 +/- 0.54, n = 10, P < 0.05) in the concentration-effect curve of endothine-1, and a right shift with smaller pD2 (from 8.10 +/- 0.23 to 7.69 +/- 0.08, n = 10, P < 0.05) in the concentration-effect curve of isoprenaline. It was suggested that in rat BSMCs there may be two types of K+ channels, Kv and BKca, which serve distinct roles. Kv participates in the control of resting Em and tension. BKca is involved in the regulation of relaxation or contraction associated with excitation.
Collapse
Affiliation(s)
- Xiansheng Liu
- Department of Respiratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | | | | | | |
Collapse
|
29
|
Oonuma H, Iwasawa K, Iida H, Nagata T, Imuta H, Morita Y, Yamamoto K, Nagai R, Omata M, Nakajima T. Inward rectifier K(+) current in human bronchial smooth muscle cells: inhibition with antisense oligonucleotides targeted to Kir2.1 mRNA. Am J Respir Cell Mol Biol 2002; 26:371-9. [PMID: 11867346 DOI: 10.1165/ajrcmb.26.3.4542] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inward rectifier K(+) (Kir) channels play an important role in forming membrane potential and then modulating muscle tone in certain types of smooth muscles. In cultured human bronchial smooth muscle cells (hBSMCs), Kir current was identified using whole-cell voltage clamp techniques and explored by using RT-PCR analysis of mRNA, Western blotting, and antisense oligonucleotide methods to block the synthesis of Kir channel protein. The K(+) current with strong inward rectification and high K(+) ion selectivity was observed. The current was unaffected by 4-aminopyridine, glibenclamide, and charybdotoxin, and hardly inhibited by tetraethylammonium, but was potently inhibited by extracellular Ba(2+). The IC(50) value of external Ba(2+) was approximately 1.3 microm. RT-PCR analysis of mRNA showed transcripts for Kir2.1, but not Kir1.1, Kir2.2, or Kir2.3. Treatment of cells with antisense oligonucleotides targeted to Kir2.1 resulted in a decrease in the current density of the Kir current and Kir protein expression, as compared with the mismatch-treated cells, whereas the current density of 4-AP-sensitive K(+) currents (K(V)) remained unaffected. The application of Ba(2+) markedly depolarized the membrane. These results demonstrate that Kir channel is present in human bronchial smooth muscle cells, and the Kir2.1 gene encodes the Kir channel protein in these cells.
Collapse
Affiliation(s)
- Hitoshi Oonuma
- Department of Respiratory Medicine, University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Potassium channel dysfunction plays a role in the pathogenesis of a number of vascular diseases including pulmonary and systemic hypertension, diabetes, and complications of atherosclerosis. Two types of K+ channels that are known to be prevalent and contribute significantly to the repolarization of vascular smooth muscle cell (SMC) membranes are the high-conductance Ca(2+)- and voltage-activated K+ (BKCa) channels, and the voltage-gated K+ (KV) channels. Alterations in either BKCa or KV channel function can have dramatic effects on vascular tone. To date, hereditary and congenital mutations in genes encoding K+ channels, abnormalities in transcription, posttranslational modifications, and altered responses to intracellular second messengers have been described as potential mechanisms for several cardiovascular diseases. Comprehensive approaches including genetic, biochemical, molecular biological, and electrophysiological analyses are necessary to identify the levels at which K+ channel expression patterns or function are disrupted. Additionally, reproducing clinical pathologies in animal, organ, and virtual models has been important in studying the discrete mechanisms by which the structure and function of these channels are altered in pathophysiological conditions. This article will describe approaches that are currently used to identify abnormalities in BKCa and KV channels that may exist in diseases involving vascular dysfunction.
Collapse
MESH Headings
- Animals
- Cardiovascular Diseases/physiopathology
- Gene Expression Regulation
- Humans
- Large-Conductance Calcium-Activated Potassium Channels
- Membrane Potentials/drug effects
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Patch-Clamp Techniques
- Potassium Channel Blockers/pharmacology
- Potassium Channels, Calcium-Activated/antagonists & inhibitors
- Potassium Channels, Calcium-Activated/genetics
- Potassium Channels, Calcium-Activated/physiology
- Potassium Channels, Voltage-Gated/antagonists & inhibitors
- Potassium Channels, Voltage-Gated/genetics
- Potassium Channels, Voltage-Gated/physiology
- Vasodilation/drug effects
- Vasodilation/physiology
Collapse
Affiliation(s)
- Victoria P Korovkina
- Department of Physiology and Biophysics, 6-432 Bowen Science Building, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
31
|
Abstract
Altered function of K+ channels associated with hypertension has been inferred from the effects of K+ channel blockers on contraction of arterial smooth muscle cells (SMCs) and from K+ efflux measurements. Of the classes of K+ channels known to exist in the smooth muscle, the contribution of voltage-gated (KV) and high-conductance, Ca2+ gated K+ (BKCa) channels to the regulation of arterial SMC contractile function has been the most studied in hypertension. The effects of selective and nonselective K+ channel blockers on tonic contraction suggest that these two K+ channel gene families contribute differently to total K+ conductance in arterial SMCs from normal and hypertensive subjects. Direct measurements of K+ channel properties by electrophysiological methods generally support this conclusion. Studies have demonstrated larger BKCa currents in SMCs from several arteries of hypertensive rats, which have been reported to result from a greater Ca2+ sensitivity of BKCa channels and/or from greater protein expression. Some, but not all, studies have shown decreased KV currents in arterial SMCs from hypertensive animals measured under Ca(2+)-replete conditions. However, when external Ca2+ is removed or when Ca2+ influx is inhibited, KV currents are larger in SMCs exposed to chronic hypertension. Gene expression studies of Shaker KV1 transcripts have shown that of the dominant species present in arterial SMCs, KV1.2 expression is higher, whereas KV1.5 is the same in SMCs from hypertensive compared to normal animals. This finding is consistent with the larger KV currents in vascular SMCs from hypertensive animals under low Ca2+ conditions and suggests that Ca2+ influx and/or intracellular Ca2+ per se exerts a greater inhibitory effect on KV currents in the myocytes from these animals. The pathways by which these K+ channel differences are produced during hypertension remain to be elucidated, as does the potential for these channel proteins to be targeted by novel antihypertensive therapies.
Collapse
Affiliation(s)
- Robert H Cox
- Lankenau Institute for Medical Research, Jefferson Health System, 100 West Lancaster Avenue, Wynnewood, Philadelphia, PA 19096, USA.
| |
Collapse
|
32
|
Thorneloe KS, Chen TT, Kerr PM, Grier EF, Horowitz B, Cole WC, Walsh MP. Molecular composition of 4-aminopyridine-sensitive voltage-gated K(+) channels of vascular smooth muscle. Circ Res 2001; 89:1030-7. [PMID: 11717160 DOI: 10.1161/hh2301.100817] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Voltage-gated K(+) channels (Kv) play a critical role in regulating arterial tone by modulating the membrane potential of vascular smooth muscle cells. Our previous work demonstrated that the dominant 4-aminopyridine (4-AP)-sensitive, delayed rectifier Kv current of rabbit portal vein (RPV) myocytes demonstrates similar 4-AP sensitivity and biophysical properties to Kv1alpha-containing channels. To identify the molecular constituents underlying the 4-AP-sensitive Kv current of vascular myocytes, we characterized the expression pattern of Kv1alpha subunits and their modulatory Kvbeta subunits in RPV. The mRNAs encoding pore-forming subunits Kv1.2, Kv1.4, and Kv1.5 were detected by reverse transcriptase-polymerase chain reaction (RT-PCR), whereas Kv1.1, Kv1.3, and Kv1.6 transcripts were undetectable. Kvbeta1.1, beta1.2, beta1.3, beta2.1, and beta2.2 messages were expressed, whereas Kvbeta3.1 and beta4 mRNAs were undetected by RT-PCR. Kv1.2, Kv1.4, Kv1.5, Kvbeta1.2, beta1.3, and beta2.1 proteins were detected in RPV by Western blotting and/or immunocytochemistry of freshly isolated myocytes. We provide the first evidence, from coimmunoprecipitation studies, for the formation of heteromultimeric Kv channel complexes composed of Kv1.2, Kv1.5, and Kvbeta1.2 subunits in vascular smooth muscle.
Collapse
Affiliation(s)
- K S Thorneloe
- Smooth Muscle Research Group, Canadian Institutes of Health Research (CIHR) Group in Regulation of Vascular Contractility, University of Calgary, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
33
|
Kurata HT, Soon GS, Fedida D. Altered state dependence of c-type inactivation in the long and short forms of human Kv1.5. J Gen Physiol 2001; 118:315-32. [PMID: 11524461 PMCID: PMC2229503 DOI: 10.1085/jgp.118.3.315] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Evidence from both human and murine cardiomyocytes suggests that truncated isoforms of Kv1.5 can be expressed in vivo. Using whole-cell patch-clamp recordings, we have characterized the activation and inactivation properties of Kv1.5DeltaN209, a naturally occurring short form of human Kv1.5 that lacks roughly 75% of the T1 domain. When expressed in HEK 293 cells, this truncated channel exhibited a V(1/2) of -19.5 +/- 0.9 mV for activation and -35.7 +/- 0.7 mV for inactivation, compared with a V(1/2) of -11.2 +/- 0.3 mV for activation and -0.9 +/- 1.6 mV for inactivation in full-length Kv.15. Kv1.5DeltaN209 channels exhibited several features rarely observed in voltage-gated K(+) channels and absent in full-length Kv1.5, including a U-shaped voltage dependence of inactivation and "excessive cumulative inactivation," in which a train of repetitive depolarizations resulted in greater inactivation than a continuous pulse. Kv1.5DeltaN209 also exhibited a stronger voltage dependence to recovery from inactivation, with the time to half-recovery changing e-fold over 30 mV compared with 66 mV in full-length Kv1.5. During trains of human action potential voltage clamps, Kv1.5DeltaN209 showed 30-35% greater accumulated inactivation than full-length Kv1.5. These results can be explained with a model based on an allosteric model of inactivation in Kv2.1 (Klemic, K.G., C.-C. Shieh, G.E. Kirsch, and S.W. Jones. 1998. Biophys. J. 74:1779-1789) in which an absence of the NH(2) terminus results in accelerated inactivation from closed states relative to full-length Kv1.5. We suggest that differential expression of isoforms of Kv1.5 may contribute to K(+) current diversity in human heart and many other tissues.
Collapse
Affiliation(s)
- Harley T. Kurata
- Department of Physiology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Gordon S. Soon
- Department of Physiology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - David Fedida
- Department of Physiology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| |
Collapse
|
34
|
Cox RH, Folander K, Swanson R. Differential Expression of Voltage-Gated K
+
Channel Genes in Arteries From Spontaneously Hypertensive and Wistar-Kyoto Rats. Hypertension 2001; 37:1315-22. [PMID: 11358947 DOI: 10.1161/01.hyp.37.5.1315] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
—Voltage-gated K
+
currents play an important role in determining membrane potential, intracellular Ca
2+
, and contraction in arterial smooth muscle. In this study, the expression of genes encoding voltage-gated K
+
channels of the Kv1.X family was compared in arteries from spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY). Expression of Kv1.X in thoracic aorta, mesenteric arteries, tail artery, and heart was determined, both qualitatively and quantitatively, by reverse transcription–polymerase chain reaction. Our results demonstrate distinct but overlapping patterns of expression in vascular tissues. In general, Kv1.2 and Kv1.5 were most highly represented, and the levels of Kv1.2 were significantly larger in all tissues from SHR. Levels of Kv1.5 in arteries did not differ significantly between strains but were greater in SHR heart. Moderate levels of Kv1.3 and Kvβ1.1 expression were also found in all tissues and were larger in SHR. Kv1.1 expression was not different between the 2 strains, and no significant expression of Kv1.4 (except in heart and aorta), Kv1.6, or Kvβ2.1 was observed in either strain. Kv1.2 and Kv1.5 transcripts represent ≈1 to 2 parts/10
5
of total mesenteric arterial RNA with ≈2- to 5-fold lower levels in aorta and tail artery. Whole-cell voltage-gated K
+
channel currents, recorded from mesenteric arterial myocytes, were larger in SHR than WKY (eg, at 0 mV: 7.3±0.8 versus 10.9±1.2 pA/pF). The voltage dependence of activation was more negative in SHR (V
0.5
: −20±4 mV versus −32±3 mV) but that of availability was not different. These results indicate that Kv1.X genes are differentially expressed between WKY and SHR (especially Kv1.2 and Kvβ1.1). These differences in gene expression are associated with a greater voltage-gated K
+
channel current density in SHR and shifted voltage-dependent activation compared with WKY. These differences may be a compensatory mechanism related to the membrane potential depolarization in SHR or some manifestation thereof.
Collapse
Affiliation(s)
- R H Cox
- Department of Physiology, University of Pennsylvania, Philadelphia, USA
| | | | | |
Collapse
|
35
|
Archer SL, Weir EK, Reeve HL, Michelakis E. Molecular identification of O2 sensors and O2-sensitive potassium channels in the pulmonary circulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 475:219-40. [PMID: 10849663 DOI: 10.1007/0-306-46825-5_21] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Small, muscular pulmonary arteries (PAs) constrict within seconds of the onset of alveolar hypoxia, diverting blood flow to better-ventilated lobes, thereby matching ventilation to perfusion and optimizing systemic PO2. This hypoxic pulmonary vasoconstriction (HPV) is enhanced by endothelial derived vasoconstrictors, such as endothelin, and inhibited by endothelial derived nitric oxide. However, the essence of the response is intrinsic to PA smooth muscle cells in resistance arteries (PASMCs). HPV is initiated by inhibition of the Kv channels in PASMCs which set the membrane potential (EM). It is currently uncertain whether this reflects an initial inhibitory effect of hypoxia on the K+ channels or an initial release of intracellular Ca2+, which then inhibits K+ channels. In either scenario, the resulting depolarization activates L-type, voltage gated Ca2+ channels, which raises cytosolic calcium levels [Ca2+]i and causes vasoconstriction. Nine families of Kv channels are recognized from cloning studies (Kv1-Kv9), each with subtypes (i.e. Kv1.1-1.6). The contribution of an individual Kv channel to the whole-cell current (IK) is difficult to determine pharmacologically because Kv channel inhibitors are nonspecific. Furthermore, the PASMC's IK is an ensemble, reflecting activity of several channels. The K+ channels which set EM, and inhibition of which initiates HPV, conduct an outward current which is slowly inactivating, and which is blocked by the Kv inhibitor 4-aminopyridine (4-AP) but not by inhibitors of Ca(2+)- or ATP-sensitive K+ channels. Using anti-Kv antibodies to immunolocalize and inhibit Kv channels, we showed that the PASMC contains numerous types of Kv channels from the Kv1 and Kv2 families., Furthermore Kv1.5 and Kv2.1 may be important in determining the EM and play a role as effectors of HPV in PASMCs. While the Kv channels in PASMCs are the "effectors" of HPV, it is uncertain whether they are intrinsically O2-sensitive or are under the control of an "O2 sensor". Certain Kv channels are rich in cysteine, and respond to the local redox environment, tending to open when oxidized and close when reduced. Candidate sensors vary the PASMC redox potential in proportion to O2. These include Nicotinamide Adenine Dinucleotide Phosphate Oxidase, (NADPH oxidase) and the cytosolic ratio of reduced/oxidized redox couples (i.e. glutathione GSH/GSSG), as controlled by electron flux in the mitochondrial electron transport chain (ETC). Using a mouse that lacks the gp91phox component of NADPH oxidase, we have recently shown that loss of the gp91phox-containing NADPH oxidase as a source of activated oxygen species does not impair HPV. However, inhibition of complex 1 of the mitochondrial electron transport chain mimics hypoxia in that it inhibits IK, reduces the production of activated O2 species and causes vasoconstriction. We hypothesize that a redox O2 sensor, perhaps in the mitochondrion, senses O2 through changes in the accumulation of freely diffusible electron donors. Changes in the ratio of reduced/oxidized redox couples, such as NADH/NAD+ and glutathione (GSH/GSSG) can reduce or oxidize the K+ channels, resulting in alterations of PA tone.
Collapse
Affiliation(s)
- S L Archer
- Department of Medicine and Physiology, University of Alberta, Canada
| | | | | | | |
Collapse
|
36
|
Oshiro T, Sasaki T, Nara M, Tamada T, Shimura S, Maruyama Y, Shirato K. Suppression of maxi-K channel and membrane depolarization by synthetic polycations in single tracheal myocytes. Am J Respir Cell Mol Biol 2000; 22:528-34. [PMID: 10783123 DOI: 10.1165/ajrcmb.22.5.3837] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Polycationic proteins, e.g., major basic protein from eosinophils or cathepsin G from neutrophils, have been shown to increase nonspecific airway responsiveness. Along with several indirect manners of action, polycations were reported to contract smooth-muscle strips and to raise the cellular Ca(2+) concentration as a direct action on airway smooth muscle. However, the mechanistic basis for the direct behavior remains to be elucidated. To address this issue, we examined the effects of synthetic cationic polypeptides poly-L-arginine and poly-L-lysine on fresh single smooth-muscle cells from bovine trachea using a patch-clamp technique. Both of the polycations significantly depolarized the membrane from a baseline of about -40 to -20 mV in a dose-dependent manner. The polycations also suppressed whole-cell spontaneous transient outward currents as well as both the conductance (from a baseline of about 130 to 70 pS) and open-state probability (about 25% of control values) of large-conductance Ca(2+)-dependent K(+) channel (maxi-K channel) on excised outside-out patch membranes. The polycations were without effect on the whole-cell Ca(2+) currents induced by depolarizing voltage pulses. We concluded that the synthetic polycations had at least two sites of action; one is the delayed rectifier K(+) channel that is responsible for the membrane depolarization that increases Ca(2+) influx, and the other is the maxi-K channel the suppression of which inhibits muscle relaxation. These results may explain the direct contractile action and, therefore, one of the mechanisms underlying the airway hyperresponsiveness induced by various polycationic proteins.
Collapse
Affiliation(s)
- T Oshiro
- First Department of Internal Medicine; and Department of Cell Physiology, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Properties, regulation, and role of potassium channels of smooth muscle. ACTA ACUST UNITED AC 2000. [DOI: 10.1016/s1569-2590(00)08010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
38
|
Shimizu S, Yokoshiki H, Sperelakis N, Paul RJ. Role of voltage-dependent and Ca(2+)-activated K(+) channels on the regulation of isometric force in porcine coronary artery. J Vasc Res 2000; 37:16-25. [PMID: 10720882 DOI: 10.1159/000025709] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We investigated the role of K(+) channels in the regulation of vascular tone in de-endothelialized porcine coronary artery. Isometric force and intracellular Ca(2+) ([Ca(2+)](i)) under resting conditions were increased by treatment with 4-aminopyridine (4-AP, 1 mM), an inhibitor of voltage-dependent K(+) (K(v)) channels, but not by tetraethylammonium chloride (TEA, 1 mM) or charybdotoxin (100 nM), both inhibitors of Ca(2+)-activated K(+) (K(Ca)) channels, or glibenclamide (10 microM), an inhibitor of ATP-sensitive K(+) channels. Under stimulated conditions with 9,11-dideoxy-11alpha, 9alpha-epoxymethano-prostaglandin F(2alpha) (U46619), 4-AP as well as TEA or charybdotoxin increased isometric force and [Ca(2+)](i), but not glibenclamide. 4-AP was the most potent in terms of depolarization of membrane potential compared with TEA or glibenclamide in the presence or absence of EGTA. In the presence of U46619, a high concentration of 4-AP (10 mM) caused a further contraction with oscillations. The force oscillations induced by 4-AP were inhibited by diltiazem (10 microM), an inhibitor of voltage-dependent Ca(2+) channels, or TEA (1 mM), but not by glibenclamide (10 microM). These force oscillations may be associated with the periodic activation of K(Ca) channels. These findings suggested that 4-AP-sensitive K(v) channels play an important role in the control of vascular tone in both resting and stimulated conditions. Moreover, under stimulated conditions, K(Ca) channels also have an important role in the regulation of vascular tone. Dysfunction of these channels induces abnormal vasoconstriction and may be implicated in vascular diseases such as hypertension and vasospasm.
Collapse
Affiliation(s)
- S Shimizu
- Department of Molecular and Cellular Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45267-0576, USA
| | | | | | | |
Collapse
|
39
|
Wade GR, Laurier LG, Preiksaitis HG, Sims SM. Delayed rectifier and Ca(2+)-dependent K(+) currents in human esophagus: roles in regulating muscle contraction. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:G885-95. [PMID: 10516156 DOI: 10.1152/ajpgi.1999.277.4.g885] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have examined K(+) channels and their function in human esophageal smooth muscle using perforated patch recording, RT-PCR to identify channel mRNA, and muscle contraction to study the effects of channel blockers. Depolarization revealed at least two types of currents: a 4-aminopyridine (4-AP)-sensitive transient delayed rectifier K(+) (K(V)) and a Ca(2+)-dependent K(+) (K(Ca)) current. K(Ca) current was active at positive potentials and was blocked by tetraethylammonium (TEA), iberiotoxin, and charybdotoxin but was insensitive to 4-AP. The mRNA encoding the gene products of Kv1.2 and Kv1.5 was identified in muscle and dissociated cells, consistent with these channel types contributing to K(V) current. 4-AP increased resting tension of muscle strips, suggesting a role for K(V) in setting the membrane potential. TEA, but not 4-AP, augmented the amplitude and duration of electrically evoked contraction, effects that were abolished by nifedipine. Here we provide the first description of macroscopic K(+) currents in human esophagus. K(V) channels participate in regulation of resting tension, whereas the K(Ca) channel limits depolarization and contraction during excitation.
Collapse
Affiliation(s)
- G R Wade
- Department of Physiology, The University of Western Ontario, Canada
| | | | | | | |
Collapse
|
40
|
Dumoulin M, Salvail D, Gaudreault SB, Cadieux A, Rousseau E. Epoxyeicosatrienoic acids relax airway smooth muscles and directly activate reconstituted KCa channels. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:L423-31. [PMID: 9728036 DOI: 10.1152/ajplung.1998.275.3.l423] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) relax various smooth muscles by increasing outward K+ movement, but the molecular mode of action of EET regioisomers remains to be clarified. The effects of EETs were investigated on bovine airway smooth muscle tone and on reconstituted Ca2+-activated K+ (KCa) channels. 5,6-EET and 11, 12-EET induced dose-dependent relaxations of precontracted bronchial spirals. These effects were partly abolished by 10 nM iberiotoxin. Bilayer experiments have shown that 0.1-10 microM 11,12-EET produced up to fourfold increases in the open probability of KCa channels from the cis (extracellular) side by enhancing the mean open time constant and reducing the long closed time constant, without affecting the unitary conductance. EET-induced activations were blocked by 10 nM iberiotoxin. Addition of vehicles or other lipids as well as of GTP and guanosine 5'-O-(3-thiotriphosphate) in the absence of EET had no effect on channel activity. Thus EETs directly activate KCa channels from airway smooth muscle through an interaction with the extracellular face of the channel. We propose that EETs could represent candidate molecules as epithelium-derived hyperpolarizing factors.
Collapse
Affiliation(s)
- M Dumoulin
- Le Bilarium, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | | | | | | | | |
Collapse
|
41
|
Waldron GJ, Sigurdsson SB, Aiello EA, Halayko AJ, Stephens NL, Cole WC. Delayed rectifier K+ current of dog bronchial myocytes: effect of pollen sensitization and PKC activation. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:L336-47. [PMID: 9700095 DOI: 10.1152/ajplung.1998.275.2.l336] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The properties of delayed rectifier K+ current [IK(dr)] of canine airway smooth muscle cells isolated from small bronchi and its modulation by protein kinase C (PKC) were studied by whole cell patch clamp. IK(dr) activated positive to -40 mV, with half-maximal activation at -16 +/- 1.2 mV (n = 15) and average current density of 31 +/- 2.6 pA/pF (n = 15) at +30 mV. The capacitive surface area, current density, and voltage dependence of activation of IK(dr) of myocytes of ragweed pollen-sensitized dogs were not different from age-matched control dogs. However, the sensitization reduced the availability of IK(dr) between -40 and -20 mV due to a hyperpolarizing shift in the voltage dependence of steady-state inactivation (-29.9 +/- 1.2 in sensitized versus -26.0 +/- 0.7 mV in control dogs, n = 9 and 11, respectively; P < 0.05). PKC activation with diacylglycerol analog or phorbol ester depressed IK(dr) amplitude, whereas an inactive diacylglycerol analog had no effect. The hyperpolarizing shift in voltage dependence of inactivation and/or modulation of IK(dr) by PKC may be two mechanisms that contribute to the enhanced reactivity of bronchial tissues from ragweed pollen-sensitized dogs.
Collapse
Affiliation(s)
- G J Waldron
- Smooth Muscle Research Group, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1
| | | | | | | | | | | |
Collapse
|
42
|
Kuriyama H, Kitamura K, Itoh T, Inoue R. Physiological features of visceral smooth muscle cells, with special reference to receptors and ion channels. Physiol Rev 1998; 78:811-920. [PMID: 9674696 DOI: 10.1152/physrev.1998.78.3.811] [Citation(s) in RCA: 176] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Visceral smooth muscle cells (VSMC) play an essential role, through changes in their contraction-relaxation cycle, in the maintenance of homeostasis in biological systems. The features of these cells differ markedly by tissue and by species; moreover, there are often regional differences within a given tissue. The biophysical features used to investigate ion channels in VSMC have progressed from the original extracellular recording methods (large electrode, single or double sucrose gap methods), to the intracellular (microelectrode) recording method, and then to methods for recording from membrane fractions (patch-clamp, including cell-attached patch-clamp, methods). Remarkable advances are now being made thanks to the application of these more modern biophysical procedures and to the development of techniques in molecular biology. Even so, we still have much to learn about the physiological features of these channels and about their contribution to the activity of both cell and tissue. In this review, we take a detailed look at ion channels in VSMC and at receptor-operated ion channels in particular; we look at their interaction with the contraction-relaxation cycle in individual VSMC and especially at the way in which their activity is related to Ca2+ movements and Ca2+ homeostasis in the cell. In sections II and III, we discuss research findings mainly derived from the use of the microelectrode, although we also introduce work done using the patch-clamp procedure. These sections cover work on the electrical activity of VSMC membranes (sect. II) and on neuromuscular transmission (sect. III). In sections IV and V, we discuss work done, using the patch-clamp procedure, on individual ion channels (Na+, Ca2+, K+, and Cl-; sect. IV) and on various types of receptor-operated ion channels (with or without coupled GTP-binding proteins and voltage dependent and independent; sect. V). In sect. VI, we look at work done on the role of Ca2+ in VSMC using the patch-clamp procedure, biochemical procedures, measurements of Ca2+ transients, and Ca2+ sensitivity of contractile proteins of VSMC. We discuss the way in which Ca2+ mobilization occurs after membrane activation (Ca2+ influx and efflux through the surface membrane, Ca2+ release from and uptake into the sarcoplasmic reticulum, and dynamic changes in Ca2+ within the cytosol). In this article, we make only limited reference to vascular smooth muscle research, since we reviewed the features of ion channels in vascular tissues only recently.
Collapse
Affiliation(s)
- H Kuriyama
- Seinan Jogakuin University, Kokura-Kita, Fukuoka, Japan
| | | | | | | |
Collapse
|
43
|
Archer SL, Souil E, Dinh-Xuan AT, Schremmer B, Mercier JC, El Yaagoubi A, Nguyen-Huu L, Reeve HL, Hampl V. Molecular identification of the role of voltage-gated K+ channels, Kv1.5 and Kv2.1, in hypoxic pulmonary vasoconstriction and control of resting membrane potential in rat pulmonary artery myocytes. J Clin Invest 1998; 101:2319-30. [PMID: 9616203 PMCID: PMC508821 DOI: 10.1172/jci333] [Citation(s) in RCA: 302] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia initiates pulmonary vasoconstriction (HPV) by inhibiting one or more voltage-gated potassium channels (Kv) in the pulmonary artery smooth muscle cells (PASMCs) of resistance arteries. The resulting membrane depolarization increases opening of voltage-gated calcium channels, raising cytosolic Ca2+ and initiating HPV. There are presently nine families of Kv channels known and pharmacological inhibitors lack the specificity to distinguish those involved in control of resting membrane potential (Em) or HPV. However, the Kv channels involved in Em and HPV have characteristic electrophysiological and pharmacological properties which suggest their molecular identity. They are slowly inactivating, delayed rectifier currents, inhibited by 4-aminopyridine (4-AP) but insensitive to charybdotoxin. Candidate Kv channels with these traits (Kv1.5 and Kv2.1) were studied. Antibodies were used to immunolocalize and functionally characterize the contribution of Kv1. 5 and Kv2.1 to PASMC electrophysiology and vascular tone. Immunoblotting confirmed the presence of Kv1.1, 1.2, 1.3, 1.5, 1.6, and 2.1, but not Kv1.4, in PASMCs. Intracellular administration of anti-Kv2.1 inhibited whole cell K+ current (IK) and depolarized Em. Anti-Kv2.1 also elevated resting tension and diminished 4-AP-induced vasoconstriction in membrane-permeabilized pulmonary artery rings. Anti-Kv1.5 inhibited IK and selectively reduced the rise in [Ca2+]i and constriction caused by hypoxia and 4-AP. However, anti-Kv1.5 neither caused depolarization nor elevated basal pulmonary artery tone. This study demonstrates that antibodies can be used to dissect the whole cell K+ currents in mammalian cells. We conclude that Kv2. 1 is an important determinant of resting Em in PASMCs from resistance arteries. Both Kv2.1 and Kv1.5 contribute to the initiation of HPV.
Collapse
Affiliation(s)
- S L Archer
- Veteran's Affairs Medical Center, Minneapolis, Minnesota 55417 and University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yuan XJ, Wang J, Juhaszova M, Golovina VA, Rubin LJ. Molecular basis and function of voltage-gated K+ channels in pulmonary arterial smooth muscle cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:L621-35. [PMID: 9575881 DOI: 10.1152/ajplung.1998.274.4.l621] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
K(+)-channel activity-mediated alteration of the membrane potential and cytoplasmic free Ca2+ concentration ([Ca2+]cyt) is a pivotal mechanism in controlling pulmonary vasomotor tone. By using combined approaches of patch clamp, imaging fluorescent microscopy, and molecular biology, we examined the electrophysiological properties of K+ channels and the role of different K+ currents in regulating [Ca2+]cyt and explored the molecular identification of voltage-gated K+ (KV)- and Ca(2+)-activated K+ (KCa)-channel genes expressed in pulmonary arterial smooth muscle cells (PASMC). Two kinetically distinct KV currents [IK(V)], a rapidly inactivating (A-type) and a noninactivating delayed rectifier, as well as a slowly activated KCa current [IK(Ca)] were identified. IK(V) was reversibly inhibited by 4-aminopyridine (5 mM), whereas IK(Ca) was significantly inhibited by charybdotoxin (10-20 nM). K+ channels are composed of pore-forming alpha-subunits and auxiliary beta-subunits. Five KV-channel alpha-subunit genes from the Shaker subfamily (KV1.1, KV1.2, KV1.4, KV1.5, and KV1.6), a KV-channel alpha-subunit gene from the Shab subfamily (KV2.1), a KV-channel modulatory alpha-subunit (KV9.3), and a KCa-channel alpha-subunit gene (rSlo), as well as three KV-channel beta-subunit genes (KV beta 1.1, KV beta 2, and KV beta 3) are expressed in PASMC. The data suggest that 1) native K+ channels in PASMC are encoded by multiple genes; 2) the delayed rectifier IK(V) may be generated by the KV1.1, KV1.2, KV1.5, KV1.6, KV2.1, and/or KV2.1/KV9.3 channels; 3) the A-type IK(V) may be generated by the KV1.4 channel and/or the delayed rectifier KV channels (KV1 subfamily) associated with beta-subunits; and 4) the IK(Ca) may be generated by the rSlo gene product. The function of the KV channels plays an important role in the regulation of membrane potential and [Ca2+]cyt in PASMC.
Collapse
Affiliation(s)
- X J Yuan
- Department of Medicine, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | | | | | |
Collapse
|
45
|
Fedida D, Chen FSP, Zhang X. The 1997 Stevenson Award Lecture. Cardiac K+channel gating: cloned delayed rectifier mechanisms and drug modulation. Can J Physiol Pharmacol 1998. [DOI: 10.1139/y98-029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
K+ channels are ubiquitous membrane proteins, which have a central role in the control of cell excitability. In the heart, voltage-gated delayed rectifier K+ channels, like Kv1.5, determine repolarization and the cardiac action potential plateau duration. Here we review the broader properties of cloned voltage-gated K+ channels with specific reference to the hKv1.5 channel in heart. We discuss the basic structural components of K+ channels such as the pore, voltage sensor, and fast inactivation, all of which have been extensively studied. Slow, or C-type, inactivation and the structural features that control pore opening are less well understood, although recent studies have given new insight into these problems. Information about channel transitions that occur prior to opening is provided by gating currents, which reflect charge-carrying transitions between kinetic closed states. By studying modulation of the gating properties of K+ channels by cations and with drugs, we can make a more complete interpretation of the state dependence of drug and ion interactions with the channel. In this way we can uncover the detailed mechanisms of action of K+ channel blockers such as tetraethylammonium ions and 4-aminopyridine, and antiarrhythmic agents such as nifedipine and quinidine.Key words: potassium channel, Kv1.5, channel gating, inactivation, pore region, gating currents.
Collapse
|
46
|
Abstract
Many important cellular functions are regulated by vascular potassium channels, including the resting membrane potential. Recent evidence suggests that the function of these channels is altered in pathophysiological disorders of the cardiovascular system. Using molecular cloning techniques, considerable effort has been made over the past 5 years to elucidate the structure of various types of potassium channels. Several different potassium channel clones have been identified from neuronal and cardiac tissues, although only a few have so far been identified in smooth muscle.
Collapse
Affiliation(s)
- L H Clapp
- Department of Medicine and Cruciform Project, Rayne Institute, University College, London, UK.
| | | |
Collapse
|
47
|
Wang J, Juhaszova M, Rubin LJ, Yuan XJ. Hypoxia inhibits gene expression of voltage-gated K+ channel alpha subunits in pulmonary artery smooth muscle cells. J Clin Invest 1997; 100:2347-53. [PMID: 9410914 PMCID: PMC508432 DOI: 10.1172/jci119774] [Citation(s) in RCA: 195] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Activity of voltage-gated K+ channels (KV) in pulmonary arterial smooth muscle cells (PASMC) is pivotal in controlling membrane potential, cytoplasmic free Ca2+ concentration ([Ca2+]cyt, and pulmonary vasomotor tone. Acute hypoxia selectively inhibits KV channels, depolarizes PASMC, raises [Ca2+]cyt, and causes pulmonary vasoconstriction and vascular remodeling. Prolonged hypoxia (24-60 h) decreased significantly the mRNA levels of KV channel alpha subunits, KV1.2 and KV1.5. Consistently, the protein levels of KV1.2 and KV1.5 were also decreased significantly by hypoxia (48-72 h). Nevertheless, hypoxia affected negligibly the mRNA levels of KV channel beta subunits (KVbeta1, KVbeta2, and KVbeta3). The native K+ channels are composed of pore-forming alpha and auxiliary beta subunits. Assembly of KV beta subunits with alpha subunits confers rapid inactivation on the slowly or non-inactivating delayed rectifier KV channels. KV beta subunits also function as an open-channel blocker of KV channels. Thus, the diminished transcription and expression of KV alpha subunits may reduce the number of KV channels and decrease KC currents. Unchanged transcription of KV beta subunits may increase the fraction of the KV channel alpha subunits that are associated with beta subunits and further reduce the total KV currents. These data demonstrate a novel mechanism by which chronic hypoxia may cause pulmonary vasoconstriction and hypertension.
Collapse
Affiliation(s)
- J Wang
- Department of Medicine, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | | | |
Collapse
|