1
|
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
|
2
|
Role of Airway Smooth Muscle in Inflammation Related to Asthma and COPD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:139-172. [PMID: 33788192 DOI: 10.1007/978-3-030-63046-1_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Airway smooth muscle contributes to both contractility and inflammation in the pathophysiology of asthma and COPD. Airway smooth muscle cells can change the degree of a variety of functions, including contraction, proliferation, migration, and the secretion of inflammatory mediators (phenotype plasticity). Airflow limitation, airway hyperresponsiveness, β2-adrenergic desensitization, and airway remodeling, which are fundamental characteristic features of these diseases, are caused by phenotype changes in airway smooth muscle cells. Alterations between contractile and hyper-contractile, synthetic/proliferative phenotypes result from Ca2+ dynamics and Ca2+ sensitization. Modulation of Ca2+ dynamics through the large-conductance Ca2+-activated K+ channel/L-type voltage-dependent Ca2+ channel linkage and of Ca2+ sensitization through the RhoA/Rho-kinase pathway contributes not only to alterations in the contractile phenotype involved in airflow limitation, airway hyperresponsiveness, and β2-adrenergic desensitization but also to alteration of the synthetic/proliferative phenotype involved in airway remodeling. These Ca2+ signal pathways are also associated with synergistic effects due to allosteric modulation between β2-adrenergic agonists and muscarinic antagonists. Therefore, airway smooth muscle may be a target tissue in the therapy for these diseases. Moreover, the phenotype changing in airway smooth muscle cells with focuses on Ca2+ signaling may provide novel strategies for research and development of effective remedies against both bronchoconstriction and inflammation.
Collapse
|
3
|
Reyes-García J, Flores-Soto E, Carbajal-García A, Sommer B, Montaño LM. Maintenance of intracellular Ca2+ basal concentration in airway smooth muscle (Review). Int J Mol Med 2018; 42:2998-3008. [PMID: 30280184 PMCID: PMC6202086 DOI: 10.3892/ijmm.2018.3910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/18/2018] [Indexed: 01/07/2023] Open
Abstract
In airway smooth muscle, the intracellular basal Ca2+ concentration [b(Ca2+)i] must be tightly regulated by several mechanisms in order to maintain a proper airway patency. The b[Ca2+]i is efficiently regulated by sarcoplasmic reticulum Ca2+-ATPase 2b, plasma membrane Ca2+-ATPase 1 or 4 and by the Na+/Ca2+ exchanger. Membranal Ca2+ channels, including the L-type voltage dependent Ca2+ channel (L-VDCC), T-type voltage dependent Ca2+ channel (T-VDCC) and transient receptor potential canonical 3 (TRPC3), appear to be constitutively active under basal conditions via the action of different signaling pathways, and are responsible for Ca2+ influx to maintain b[Ca2+]i. The two types of voltage-dependent Ca2+ channels (L- and T-type) are modulated by phosphorylation processes mediated by mitogen-activated protein kinase kinase (MEK) and extracellular-signal-regulated kinase 1 and 2 (ERK1/2). The MEK/ERK signaling pathway can be activated by G-protein-coupled receptors through the αq subunit when the endogenous ligand (i.e., acetylcholine, histamine, leukotrienes, etc.) is present under basal conditions. It may also be stimulated when receptor tyrosine kinases are occupied by the appropriate ligand (cytokines, growth factors, etc.). ERK1/2 phosphorylates L-VDCC on Ser496 of the β2 subunit and Ser1928 of the α1 subunit, decreasing or increasing the channel activity, respectively, and enabling it to switch between an open and closed state. T-VDCC is also probably phosphorylated by ERK1/2, although further research is required to identify the phosphorylation sites. TRPC3 is directly activated by diacylglycerol produced by phospholipase C (PLCβ or γ). Constitutive inositol 1,4,5-trisphosphate production induces the release of Ca2+ from the sarcoplasmic reticulum through inositol triphosphate receptor 1. This ion induces Ca2+-induced Ca2+ release through the ryanodine receptor 2 (designated as Ca2+ ‘sparks’). Therefore, several Ca2+ handling mechanisms are finely tuned to regulate basal intracellular Ca2+ concentrations. It is conceivable that alterations in any of these processes may render airway smooth muscle susceptible to develop hyperresponsiveness that is observed in ailments such as asthma.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México 14080, México
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| |
Collapse
|
4
|
Zhao Z, Yin Y, Wang Z, Fang R, Wu H, Jiang M, Bai G, Luo G. Arctigenin exhibits relaxation effect on bronchus by affecting transmembrane flow of calcium. Biol Trace Elem Res 2013; 156:181-7. [PMID: 24114345 DOI: 10.1007/s12011-013-9839-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 09/26/2013] [Indexed: 11/27/2022]
Abstract
Arctigenin, a lignan extract from Arctium lappa (L.), exhibits anti-inflammation, antioxidation, vasodilator effects, etc. However, the effects of arctigenin on bronchus relaxation are not well investigated. This study aimed to investigate how arctigenin regulates bronchus tone and calcium ion (Ca(2+)) flow. Trachea strips of guinea pigs were prepared for testing the relaxation effect of arctigenin to acetylcholine, histamine, KCl, and CaCl2, respectively. Furthermore, L-type calcium channel currents were detected by patch-clamp, and intracellular Ca(2+) concentration was detected by confocal microscopy. The results showed that arctigenin exhibited relaxation effect on tracheae to different constrictors, and this was related to decreasing cytoplasmic Ca(2+) concentration by inhibiting Ca(2+) influx partly through L-type calcium channel as well as promoting Ca(2+) efflux. In summary, this study provides new insight into the mechanisms by which arctigenin exhibits relaxation effect on bronchus and suggests its potential use for airway disease therapy.
Collapse
Affiliation(s)
- Zhenying Zhao
- College of Pharmacy, Nankai University, Weijin Road 94#, Tianjin, 300071, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Vieira AB, Coelho LP, Insuela DBR, Carvalho VF, dos Santos MH, Silva PMR, Martins MA. Mangiferin prevents guinea pig tracheal contraction via activation of the nitric oxide-cyclic GMP pathway. PLoS One 2013; 8:e71759. [PMID: 23951240 PMCID: PMC3738528 DOI: 10.1371/journal.pone.0071759] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/03/2013] [Indexed: 12/26/2022] Open
Abstract
Previous studies have described the antispasmodic effect of mangiferin, a natural glucoside xanthone (2-C-β-Dgluco-pyranosyl-1,3,6,7-tetrahydroxyxanthone) that is present in mango trees and other plants, but its mechanism of action remains unknown. The aim of this study was to examine the potential contribution of the nitric oxide-cyclic GMP pathway to the antispasmodic effect of mangiferin on isolated tracheal rings preparations. The functional effect of mangiferin on allergic and non-allergic contraction of guinea pig tracheal rings was assessed in conventional organ baths. Cultured tracheal rings were exposed to mangiferin or vehicle, and nitric oxide synthase (NOS) 3 and cyclic GMP (cGMP) levels were quantified using western blotting and enzyme immunoassays, respectively. Mangiferin (0.1–10 µM) inhibited tracheal contractions induced by distinct stimuli, such as allergen, histamine, 5-hydroxytryptamine or carbachol, in a concentration-dependent manner. Mangiferin also caused marked relaxation of tracheal rings that were precontracted by carbachol, suggesting that it has both anti-contraction and relaxant properties that are prevented by removing the epithelium. The effect of mangiferin was inhibited by the nitric oxide synthase inhibitor, Nω-nitro-L-arginine methyl ester (L-NAME) (100 µM), and the soluble guanylate cyclase inhibitor, 1H-[1], [2], [4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) (10 µM), but not the adenylate cyclase inhibitor, 9-(tetrahydro-2-furyl)adenine (SQ22536) (100 µM). The antispasmodic effect of mangiferin was also sensitive to K+ channel blockers, such as tetraethylammonium (TEA), glibenclamide and apamin. Furthermore, mangiferin inhibited Ca2+-induced contractions in K+ (60 mM)-depolarised tracheal rings preparations. In addition, mangiferin increased NOS3 protein levels and cGMP intracellular levels in cultured tracheal rings. Finally, mangiferin-induced increase in cGMP levels was abrogated by co-incubation with either ODQ or L-NAME. These data suggest that the antispasmodic effect of mangiferin is mediated by epithelium-nitric oxide- and cGMP-dependent mechanisms.
Collapse
Affiliation(s)
- Aline B. Vieira
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Luciana P. Coelho
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Daniella B. R. Insuela
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Vinicius F. Carvalho
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Marcelo H. dos Santos
- Laboratory of Phytochemistry and Medicinal and Chemistry, Department of Pharmacy, Alfenas, Federal University of Alfenas, MG, Brazil
| | - Patricia MR. Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Marco A. Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
- * E-mail:
| |
Collapse
|
6
|
Molecular expression and functional role of canonical transient receptor potential channels in airway smooth muscle cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:731-47. [PMID: 21290324 DOI: 10.1007/978-94-007-0265-3_38] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple canonical or classic transient receptor potential (TRPC) molecules are expressed in animal and human airway smooth muscle cells (SMCs). TRPC3, but not TRPC1, is a major molecular component of native non-selective cation channels (NSCCs) to contribute to the resting [Ca(2+)](i) and muscarinic increase in [Ca(2+)](i) in freshly isolated airway SMCs. TRPC3-encoded NSCCs are significantly increased in expression and activity in airway SMCs from ovalbumin-sensitized/challenged "asthmatic" mice, whereas TRPC1-encoded channel activity, but not its expression, is largely augmented. The upregulated TRPC3- and TRPC1-encoded NSCC activity both mediate "asthmatic" membrane depolarization in airway SMCs. Supportively, tumor necrosis factor-α (TNFα), an important asthma mediator, increases TRPC3 expression, and TRPC3 gene silencing inhibits TNFα-mediated augmentation of acetylcholine-evoked increase in [Ca(2+)](i) in passaged airway SMCs. In contrast, TRPC6 gene silencing has no effect on 1-oleoyl-2-acetyl-sn-glycerol (OAG)-evoked increase in [Ca(2+)](i) in primary isolated cells. These findings provide compelling information indicating that TRPC3-encoded NSCCs are important for physiological and pathological cellular responses in airway SMCs. However, continual studies are necessary to further determine whether, which, and how TRPC-encoded channels are involved in cellular responses in normal and diseased (e.g., asthmatic) airway SMCs.
Collapse
|
7
|
Abstract
The sarcoplasmic reticulum (SR) of smooth muscles presents many intriguing facets and questions concerning its roles, especially as these change with development, disease, and modulation of physiological activity. The SR's function was originally perceived to be synthetic and then that of a Ca store for the contractile proteins, acting as a Ca amplification mechanism as it does in striated muscles. Gradually, as investigators have struggled to find a convincing role for Ca-induced Ca release in many smooth muscles, a role in controlling excitability has emerged. This is the Ca spark/spontaneous transient outward current coupling mechanism which reduces excitability and limits contraction. Release of SR Ca occurs in response to inositol 1,4,5-trisphosphate, Ca, and nicotinic acid adenine dinucleotide phosphate, and depletion of SR Ca can initiate Ca entry, the mechanism of which is being investigated but seems to involve Stim and Orai as found in nonexcitable cells. The contribution of the elemental Ca signals from the SR, sparks and puffs, to global Ca signals, i.e., Ca waves and oscillations, is becoming clearer but is far from established. The dynamics of SR Ca release and uptake mechanisms are reviewed along with the control of luminal Ca. We review the growing list of the SR's functions that still includes Ca storage, contraction, and relaxation but has been expanded to encompass Ca homeostasis, generating local and global Ca signals, and contributing to cellular microdomains and signaling in other organelles, including mitochondria, lysosomes, and the nucleus. For an integrated approach, a review of aspects of the SR in health and disease and during development and aging are also included. While the sheer versatility of smooth muscle makes it foolish to have a "one model fits all" approach to this subject, we have tried to synthesize conclusions wherever possible.
Collapse
Affiliation(s)
- Susan Wray
- Department of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, Merseyside L69 3BX, United Kingdom.
| | | |
Collapse
|
8
|
Coelho LP, Serra MF, Pires ALDA, Cordeiro RSB, Rodrigues e Silva PM, dos Santos MH, Martins MA. 7-Epiclusianone, a tetraprenylated benzophenone, relaxes airway smooth muscle through activation of the nitric oxide-cGMP pathway. J Pharmacol Exp Ther 2008; 327:206-14. [PMID: 18591220 DOI: 10.1124/jpet.108.138032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was undertaken to investigate the putative mechanism(s) underlying the antispasmodic effect of 7-epiclusianone, a naturally occurring compound isolated from the plant Garcinia brasiliensis. Guinea pig tracheal rings were mounted in tissue baths filled with Krebs' solution, and the contractile response to distinct stimuli was measured in the presence or absence of 7-epiclusianone. We also tested the effect of 7-epiclusianone on methacholine-evoked airways obstruction in BALB/c mice using barometric plethysmography. 7-Epiclusianone (10 microM) inhibited epithelium-intact tracheal ring contraction induced by allergen, histamine, 5-hydroxytryptamine, or carbachol challenge. The relaxation effect was abrogated by epithelium removal, the presence of nitric-oxide synthase inhibitor N(omega)-nitro-L-arginine methyl ester (L-NAME) (100 microM), or soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) (10 microM). 7-Epiclusianone (1-100 microM) induced a dose-dependent increase in the intracellular cGMP levels of cultured tracheal rings. The relaxation effect of 7-epiclusianone was also inhibited by K(+) channel blockers tetraethylammonium (10 microM), glibenclamide (1 microM), or apamin (1 microM), but not by 9-(tetrahydro-2'-furyl)adenine (SQ22,536) (100 microM), an adenylate cyclase inhibitor. In epithelium-intact tracheal rings, 7-epiclusianone also inhibited Ca(2+)-induced contractions in K(+) (60 mM)-depolarized preparations, but it seemed ineffective in assays in which epithelium-denuded tracheal ring preparations were used. Oral administration of 7-epiclusinone (25-100 mg/kg) dose-dependently inhibited airway obstruction triggered by aerosolized methacholine (6-25 mg/ml), in a mechanism sensitive to L-NAME (20 mg/kg). In conclusion, the relaxation effect of 7-epiclusinone seems to be mediated by epithelium-, nitric oxide-, and cGMP-dependent mechanisms. Furthermore, oral administration of 7-epiclusianone reduces episodes of bronchial obstruction, warranting further research on this compound regarding a putative application in asthma therapy.
Collapse
Affiliation(s)
- Luciana Pontes Coelho
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4365, Manguinhos, CEP 21045-900, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | |
Collapse
|
9
|
Liu C, Zuo J, Pertens E, Helli PB, Janssen LJ. Regulation of Rho/ROCK signaling in airway smooth muscle by membrane potential and [Ca2+]i. Am J Physiol Lung Cell Mol Physiol 2005; 289:L574-82. [PMID: 15937065 DOI: 10.1152/ajplung.00134.2005] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recently, we have shown that Rho and Rho-activated kinase (ROCK) may become activated by high-millimolar KCl, which had previously been widely assumed to act solely through opening of voltage-dependent Ca(2+) channels. In this study, we explored in more detail the relationship between membrane depolarization, Ca(2+) currents, and activation of Rho/ROCK in bovine tracheal smooth muscle. Ca(2+) currents began to activate at membrane voltages more positive than -40 mV and were maximally activated above 0 mV; at the same time, these underwent time- and voltage-dependent inactivation. Depolarizing intact tissues by KCl challenge evoked contractions that were blocked equally, and in a nonadditive fashion, by nifedipine or by the ROCK inhibitor Y-27632. Other agents that elevate intracellular calcium concentration ([Ca(2+)](i)) by pathways independent of G protein-coupled receptors, namely the SERCA-pump inhibitor cyclopiazonic acid and the Ca(2+) ionophore A-23187, evoked contractions that were also largely reduced by Y-27632. KCl directly increased Rho and ROCK activities in a concentration-dependent fashion that paralleled closely the effect of KCl on tone and [Ca(2+)](i), as well as the voltage-dependent Ca(2+) currents that were measured over the voltage ranges that are evoked by 0-120 mM KCl. Through the use of various pharmacological inhibitors, we ruled out roles for Ca(2+)/calmodulin-dependent CaM kinase II, protein kinase C, and protein kinase A in mediating the KCl-stimulated changes in tone and Rho/ROCK activities. In conclusion, Rho is activated by elevation of [Ca(2+)](i) (although the signal transduction pathway underlying this Ca(2+) dependence is still unclear) and possibly also by membrane depolarization per se.
Collapse
Affiliation(s)
- Caiqiong Liu
- Asthma Research Group, Firestone Institute for Respiratory Health, St. Joseph's Hospital, Hamilton, Ontario, Canada L8N 4A6
| | | | | | | | | |
Collapse
|
10
|
Jia Y, Wang X, Varty L, Rizzo CA, Yang R, Correll CC, Phelps PT, Egan RW, Hey JA. Functional TRPV4 channels are expressed in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2004; 287:L272-8. [PMID: 15075247 DOI: 10.1152/ajplung.00393.2003] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypotonic stimulation induces airway constriction in normal and asthmatic airways. However, the osmolarity sensor in the airway has not been characterized. TRPV4 (also known as VR-OAC, VRL-2, TRP12, OTRPC4), an osmotic-sensitive cation channel in the transient receptor potential (TRP) channel family, was recently cloned. In the present study, we show that TRPV4 mRNA was expressed in cultured human airway smooth muscle cells as analyzed by RT-PCR. Hypotonic stimulation induced Ca(2+) influx in human airway smooth muscle cells in an osmolarity-dependent manner, consistent with the reported biological activity of TRPV4 in transfected cells. In cultured muscle cells, 4alpha-phorbol 12,13-didecanoate (4-alphaPDD), a TRPV4 ligand, increased intracellular Ca(2+) level only when Ca(2+) was present in the extracellular solution. The 4-alphaPDD-induced Ca(2+) response was inhibited by ruthenium red (1 microM), a known TRPV4 inhibitor, but not by capsazepine (1 microM), a TRPV1 antagonist, indicating that 4-alphaPDD-induced Ca(2+) response is mediated by TRPV4. Verapamil (10 microM), an L-type voltage-gated Ca(2+) channel inhibitor, had no effect on the 4-alphaPDD-induced Ca(2+) response, excluding the involvement of L-type Ca(2+) channels. Furthermore, hypotonic stimulation elicited smooth muscle contraction through a mechanism dependent on membrane Ca(2+) channels in both isolated human and guinea pig airways. Hypotonicity-induced airway contraction was not inhibited by the L-type Ca(2+) channel inhibitor nifedipine (1 microM) or by the TRPV1 inhibitor capsazepine (1 microM). We conclude that functional TRPV4 is expressed in human airway smooth muscle cells and may act as an osmolarity sensor in the airway.
Collapse
Affiliation(s)
- Yanlin Jia
- Neurobiology, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA. )
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Wellman GC, Nelson MT. Signaling between SR and plasmalemma in smooth muscle: sparks and the activation of Ca2+-sensitive ion channels. Cell Calcium 2003; 34:211-29. [PMID: 12887969 DOI: 10.1016/s0143-4160(03)00124-6] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Intracellular calcium ions are involved in the regulation of nearly every aspect of cell function. In smooth muscle, Ca2+ can be delivered to Ca2+-sensitive effector molecules either by influx through plasma membrane ion channels or by intracellular Ca2+ release events. Ca2+ sparks are transient local increases in intracellular Ca2+ that arise from the opening of ryanodine-sensitive Ca2+ release channels (ryanodine receptors) located in the sarcoplasmic reticulum. In arterial myocytes, Ca2+ sparks occur near the plasma membrane and act to deliver high (microM) local Ca2+ to plasmalemmal Ca2+-sensitive ion channels, without directly altering global cytosolic Ca2+ concentrations. The two major ion channel targets of Ca2+ sparks are Ca2+-activated chloride (Cl(Ca)) channels and large-conductance Ca2+-activated potassium (BK) channels. The activation of BK channels by Ca2+ sparks play an important role in the regulation of arterial diameter and appear to be involved in the action of a variety of vasodilators. The coupling of Ca2+ sparks to BK channels can be influenced by a number of factors including membrane potential and modulatory beta subunits of BK channels. Cl(Ca) channels, while not present in all smooth muscle, can also be activated by Ca2+ sparks in some types of smooth muscle. Ca2+ sparks can also influence the activity of Ca2+-dependent transcription factors and expression of immediate early response genes such as c-fos. In summary, Ca2+ sparks are local Ca2+ signaling events that in smooth muscle can act on plasma membrane ion channels to influence excitation-contraction coupling as well as gene expression.
Collapse
Affiliation(s)
- George C Wellman
- Department of Pharmacology, The University of Vermont College of Medicine, Given Building, Room B-321, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| | | |
Collapse
|
12
|
Lee JCF, Barajas-López C, Huizinga JD. Heterogeneous expression of transient outward currents in smooth muscle cells of the mouse small intestine. J Physiol 2002; 544:19-27. [PMID: 12356877 PMCID: PMC2290574 DOI: 10.1113/jphysiol.2001.013557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The objective for this paper was to characterize the transient outward current (I(to)) present in smooth muscle cells of the intestinal external muscularis. Two populations of cells were identified, one with a fast rate of I(to) inactivation (tau < 100 ms) and another with a slow rate of I(to) inactivating (tau > 150 ms). The chord conductance for the fast I(to) was smaller than the chord conductance of the slow I(to) (0.5 +/- 0.1 vs. 1.3 +/- 0.1 nS pF(-1), respectively). The inactivation was fitted by mono-exponentials to give a tau for the fast and slow I(to) of 44 and 229 ms, respectively. Combined plots of voltage dependent activation and inactivation processes revealed voltage ranges where window currents were possible; a 20 mV range for the fast I(to) from -56 to -36 mV and a 47 mV range for the slow I(to) from -42 to +5 mV. The fast I(to) recovered more quickly from inactivation than the slow I(to); tau(fast I(to)) = 11 +/- 4 ms compared to tau(slow I(to)) = 42 +/- 16 ms. The effect of different rates of depolarization on I(to) activation was examined. The plots of peak currents evoked by different rates of depolarization were well fitted by inverse exponential functions. The fast I(to) had a larger response to fast rates of depolarization by having a tau of 2 +/- 1 mV ms(-1) with maximal activation (95 % complete) at 5 mV ms(-1). The slow I(to) had a tau of 14 +/- 9 mV ms(-1) with maximal activation (95 % complete) at 42 mV ms(-1). The properties of these currents suggest that the two transient outward currents may contribute differently to slow waves and action potentials generated by the smooth muscle cells.
Collapse
Affiliation(s)
- Jonathan C F Lee
- Intestinal Disease Research Program, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | | | | |
Collapse
|
13
|
Janssen LJ. Ionic mechanisms and Ca(2+) regulation in airway smooth muscle contraction: do the data contradict dogma? Am J Physiol Lung Cell Mol Physiol 2002; 282:L1161-78. [PMID: 12003770 DOI: 10.1152/ajplung.00452.2001] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In general, excitation-contraction coupling in muscle is dependent on membrane depolarization and hyperpolarization to regulate the opening of voltage-dependent Ca(2+) channels and, thereby, influence intracellular Ca(2+) concentration ([Ca(2+)](i)). Thus Ca(2+) channel blockers and K(+) channel openers are important tools in the arsenals against hypertension, stroke, and myocardial infarction, etc. Airway smooth muscle (ASM) also exhibits robust Ca(2+), K(+), and Cl(-) currents, and there are elaborate signaling pathways that regulate them. It is easy, then, to presume that these also play a central role in contraction/relaxation of ASM. However, several lines of evidence speak to the contrary. Also, too many researchers in the ASM field view the sarcoplasmic reticulum as being centrally located and displacing its contents uniformly throughout the cell, and they have focused almost exclusively on the initial single [Ca(2+)] spike evoked by excitatory agonists. Several recent studies have revealed complex spatial and temporal heterogeneity in [Ca(2+)](i), the significance of which is only just beginning to be appreciated. In this review, we will compare what is known about ion channels in ASM with what is believed to be their roles in ASM physiology. Also, we will examine some novel ionic mechanisms in the context of Ca(2+) handling and excitation-contraction coupling in ASM.
Collapse
Affiliation(s)
- Luke J Janssen
- Asthma Research Group, Firestone Institute for Respiratory Health, St. Joseph's Hospital, McMaster University, Hamilton, Ontario, Canada L8N 4A6.
| |
Collapse
|
14
|
Wu SN, Chen CC, Li HF, Lo YK, Chen SA, Chiang HT. Stimulation of the BK(Ca) channel in cultured smooth muscle cells of human trachea by magnolol. Thorax 2002; 57:67-74. [PMID: 11809993 PMCID: PMC1746183 DOI: 10.1136/thorax.57.1.67] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Magnolol, a compound isolated from the cortex of Magnolia officinalis, has been found to possess anti-allergic and anti-asthmatic activity. METHODS The effect of magnolol on ionic currents was studied in cultured smooth muscle cells of human trachea with the aid of the patch clamp technique. RESULTS In whole cell current recordings magnolol reversibly increased the amplitude of K+ outward currents. The increase in outward current caused by magnolol was sensitive to inhibition by iberiotoxin (200 nM) or paxilline (1 microM) but not by glibenclamide (10 microM). In inside out patches, magnolol added to the bath did not modify single channel conductance but effectively enhanced the activity of large conductance Ca2+ activated K+ (BK(Ca)) channels. Magnolol increased the probability of these channel openings in a concentration dependent manner with an EC50 value of 1.5 microM. The magnolol stimulated increase in the probability of channels opening was independent of internal Ca2+. The application of magnolol also shifted the activation curve of BK(Ca) channels to less positive membrane potentials. The change in the kinetic behaviour of BK(Ca) channels caused by magnolol in these cells is the result of an increase in dissociation and gating constants. CONCLUSIONS These results provide evidence that, in addition to the presence of antioxidative activity, magnolol is potent in stimulating BK(Ca) channel activity in tracheal smooth muscle cells. The direct stimulation of these BK(Ca) channels by magnolol may contribute to the underlying mechanism by which it acts as an anti-asthmatic compound.
Collapse
Affiliation(s)
- S-N Wu
- Department of Medical Education and Research, Kaohsiung-Veterans General Hospital, Kaohsiung City, Taiwan.
| | | | | | | | | | | |
Collapse
|
15
|
Pabelick CM, Sieck GC, Prakash YS. Invited review: significance of spatial and temporal heterogeneity of calcium transients in smooth muscle. J Appl Physiol (1985) 2001; 91:488-96. [PMID: 11408467 DOI: 10.1152/jappl.2001.91.1.488] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The multiplicity of mechanisms involved in regulation of intracellular Ca(2+) concentration ([Ca(2+)](i)) in smooth muscle results in both intra- and intercellular heterogeneities in [Ca(2+)](i). Heterogeneity in [Ca(2+)](i) regulation is reflected by the presence of spontaneous, localized [Ca(2+)](i) transients (Ca(2+) sparks) representing Ca(2+) release through ryanodine receptor (RyR) channels. Ca(2+) sparks display variable spatial Ca(2+) distributions with every occurrence within and across cellular regions. Individual sparks are often grouped, and fusion of sparks produces large local elevations in [Ca(2+)](i) that occasionally trigger propagating [Ca(2+)](i) waves. Ca(2+) sparks may modulate membrane potential and thus smooth muscle contractility. Sparks may also be the target of other regulatory factors in smooth muscle. Agonists induce propagating [Ca(2+)](i) oscillations that originate from foci with high spark incidence and also represent Ca(2+) release through RyR channels. With increasing agonist concentration, the peak of regional [Ca(2+)](i) oscillations remains relatively constant, whereas both frequency and propagation velocity increase. In contrast, the global cellular response appears as a concentration-dependent increase in peak as well as mean cellular [Ca(2+)](i), representing a spatial and temporal integration of the oscillations. The significance of agonist-induced [Ca(2+)](i) oscillations lies in the establishment of a global [Ca(2+)](i) level for slower Ca(2+)-dependent physiological processes.
Collapse
Affiliation(s)
- C M Pabelick
- Department of Anesthesiology, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
16
|
Abstract
Local intracellular Ca(2+) transients, termed Ca(2+) sparks, are caused by the coordinated opening of a cluster of ryanodine-sensitive Ca(2+) release channels in the sarcoplasmic reticulum of smooth muscle cells. Ca(2+) sparks are activated by Ca(2+) entry through dihydropyridine-sensitive voltage-dependent Ca(2+) channels, although the precise mechanisms of communication of Ca(2+) entry to Ca(2+) spark activation are not clear in smooth muscle. Ca(2+) sparks act as a positive-feedback element to increase smooth muscle contractility, directly by contributing to the global cytoplasmic Ca(2+) concentration ([Ca(2+)]) and indirectly by increasing Ca(2+) entry through membrane potential depolarization, caused by activation of Ca(2+) spark-activated Cl(-) channels. Ca(2+) sparks also have a profound negative-feedback effect on contractility by decreasing Ca(2+) entry through membrane potential hyperpolarization, caused by activation of large-conductance, Ca(2+)-sensitive K(+) channels. In this review, the roles of Ca(2+) sparks in positive- and negative-feedback regulation of smooth muscle function are explored. We also propose that frequency and amplitude modulation of Ca(2+) sparks by contractile and relaxant agents is an important mechanism to regulate smooth muscle function.
Collapse
Affiliation(s)
- J H Jaggar
- Department of Pharmacology, College of Medicine, The University of Vermont, Burlington, Vermont 05405, USA
| | | | | | | |
Collapse
|
17
|
Li L, Vaali K, Vapaatalo H, Kankaanranta H. Effects of K(+) channel inhibitors on relaxation induced by flufenamic and tolfenamic acids in guinea-pig trachea. Eur J Pharmacol 1999; 383:169-76. [PMID: 10585531 DOI: 10.1016/s0014-2999(99)00634-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effects of different K(+) channel inhibitors on flufenamic- and tolfenamic-acids-induced relaxation were studied in prostaglandin F(2alpha) (1 microM) precontracted guinea-pig trachea. Flufenamic and tolfenamic acids (each 0.1-33 microM) and lemakalim (0.01-33 microM), but not indomethacin (0.1-33 microM), caused relaxation. Iberiotoxin (33 and 100 nM) inhibited flufenamic- and tolfenamic-acids-, but not lemakalim-, induced relaxation. Iberiotoxin (100 nM) inhibited nifedipine (10 nM-10 microM)-induced relaxation. Nifedipine (0.1 microM) inhibited the blockade of fenamate-induced relaxation by iberiotoxin. Apamin (0.1 and 1 microM) did not affect flufenamic- and tolfenamic-acids- and lemakalim-induced relaxation. Glibenclamide (10 and 33 microM) inhibited lemakalim-, but not flufenamic- and tolfenamic-acids-, induced relaxation. 4-Aminopyridine (0.5 and 2 mM) inhibited flufenamic- and tolfenamic- acids- and lemakalim-induced relaxation. Flufenamic- and tolfenamic-acids-induced relaxation is likely to be activation of Ca(2+)-activated K(+) channels and differs from that of lemakalim.
Collapse
Affiliation(s)
- L Li
- Institute of Biomedicine, Department of Pharmacology and Toxicology, P.O. Box 8, University of Helsinki, Helsinki, Finland.
| | | | | | | |
Collapse
|
18
|
Yamashita T, Kokubun S. Nonselective cationic currents activated by acetylcholine in swine tracheal smooth muscle cells. Can J Physiol Pharmacol 1999. [DOI: 10.1139/y99-069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Membrane currents in isolated swine tracheal smooth muscle cells were investigated using a pipette solution containing BAPTA-Ca2+ buffer and Cs+ as the major cation. With a pipette solution containing 100 nM free Ca2+, acetylcholine (ACh; 1-100 µM), in a concentration-dependent manner, activated a current without inducing shortening of cells, although neither 1 mM histamine nor 1 µM leukotriene D4 activated the current (n = 7, n is the number of cells). The effect of 100 µM ACh was suppressed by pretreatment with 100 µM atropine (n = 6) or intracellular application of preactivated pertussis toxin at a concentration of 0.1 µg·mL-1 (n = 8). Genistein (0.1-100 µM), in a concentration-dependent manner, suppressed the activation of the inward current by 100 µM ACh, whereas it did not significantly suppress that of the outward current (n = 6-8). With a pipette solution containing 50 nM free Ca2+, outward current, but not inward current, was activated by 100 µM ACh (n = 10). When the pipette solution had free Ca2+ concentrations greater than 50 nM, the inward current together with the outward current was activated. The ratio between the amplitude of the inward and outward currents was significantly increased as the free Ca2+ concentration in the pipette solution increased. The steady-state activation curve of the ACh-activated current with the 50 nM free Ca2+ pipette solution was fitted by a single Boltzmann distribution (Vh = +69.8 mV, k = -11.9 mV, n = 10). The activation time constant became smaller as the membrane potential was more depolarized (164.3 ± 5.9 ms at +40 mV to 92.4 ± 6.3 ms at +120 mV, n = 10). The reversal potential was not significantly changed by reducing extracellular Cl- concentration to one-tenth of the control (n = 8), suggesting that the current is a nonselective cationic current. These results suggest that ACh activates an outward nonselective cationic current via pertussis toxin-sensitive G-protein(s) coupled with muscarinic receptors. Involvement of genistein-sensitive tyrosine kinase in the activation process of the current is unlikely.Key words: tracheal smooth muscle, nonselective cationic current, acetylcholine, Ca2+ dependency, genistein sensitivity.
Collapse
|
19
|
Waniishi Y, Inoue R, Morita H, Teramoto N, Abe K, Ito Y. Cyclic GMP-dependent but G-kinase-independent inhibition of Ca2+-dependent Cl- currents by NO donors in cat tracheal smooth muscle. J Physiol 1998; 511 ( Pt 3):719-31. [PMID: 9714855 PMCID: PMC2231153 DOI: 10.1111/j.1469-7793.1998.719bg.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
1. The effects of NO donors on Ca2+-dependent Cl- currents (ICl(Ca)) were investigated in freshly isolated cat tracheal myocytes using the whole-cell patch clamp technique. 2. With nystatin-perforated whole-cell recording, carbachol (CCh, >/= 1 microM) induced a transient inward current (ICCh) with a reversal potential of about -20 mV. Activation of ICCh probably occurred through the M3 muscarinic receptor, since nanomolar concentrations of 4-diphenylacetoxy-N-methylpiperidine methobromide (4-DAMP) greatly inhibited this current, while 11-(2-(diethylamino)methyl)-1-piperidinylacetyl)-5, 11-dihydro-6H-pyrido (2,3beta) (1,4)benzodiazepine-6-one (AF-DX 116) or pirenzepine at concentrations of up to 1 microM were almost ineffective. 3. Chloride channel/transporter blockers such as DIDS (100 microM), anthracene-9-carboxylic acid (9-AC, 100 microM) and niflumic acid (100 microM) greatly inhibited ICCh, but cation channel blockers, such as nifedipine (10 microM), Zn2+ (500 microM) or Gd3+ (500 microM), were without effect. 4. Activation of ICCh was strongly attenuated by pretreatment with ryanodine (4 microM) plus caffeine (10 mM). Addition of neomycin (1 mM) into the bath or inclusion of heparin (3 mg ml-1) in the pipette abolished a substantial part of ICCh. These results suggest that ICCh is ICl(Ca), which is activated by inositol 1,4,5-trisphosphate (IP3)-mediated Ca2+ release. 5. The nitric oxide donor S-nitroso-N-acetyl penicillamine (SNAP) reduced the amplitude of ICCh dose dependently (IC50, approximately 10 microM). Similar inhibition was also exerted by other types of NO donor such as glyceryl trinitrate (GTN) and (+/-)-E-methyl-2-(E-hydroxyimitol)-5-nitro-6-methoxy-3- hexeneamide (NO-R). 6. SNAP-induced ICCh inhibition was effectively antagonized by Methylene Blue (1-100 nM), and mimicked by dibutyryl cGMP (db-cGMP) (0.5-1 mM), whereas two structurally distinct types of cGMP-dependent (G)-kinase inhibitor, N-(2-aminoethyl)-5-isoquinilinesulphonamide (H-8, 2.5 microM) and KT5823 (1 microM), failed to counteract the inhibitory effects of SNAP or db-cGMP. Another G-kinase-specific inhibitor Rp-8-(para-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate (Rp-8-pCPT-cGMPS; 1 microM) itself caused a marked reduction in ICCh. 7. SNAP (100 microM) or db-cGMP (100 microM) exhibited no inhibitory actions, when caffeine (10 mM) or photolytically released IP3 were used instead of CCh to activate the inward current. 8. These results suggest that inhibition of ICCh by NO donors involves a cGMP-dependent but G-kinase-independent mechanism, which may operate at a site(s) between the muscarinic (M3) and IP3 receptors.
Collapse
Affiliation(s)
- Y Waniishi
- Department of Pharmacology, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Janssen LJ, Hague C, Nana R. Ionic mechanisms underlying electrical slow waves in canine airway smooth muscle. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:L516-23. [PMID: 9728046 DOI: 10.1152/ajplung.1998.275.3.l516] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In canine bronchial smooth muscle (BSM), spasmogens evoke oscillations in membrane potential ("slow waves"). The depolarizing phase of the slow waves is mediated by voltage-dependent Ca2+ channels; we examined the roles played by Cl- and K+ currents and Na+-K+-ATPase activity in mediating the repolarizing phase. Slow waves were evoked using tetraethylammonium (25 mM) in the presence or absence of niflumic acid (100 microM; Cl- channel blocker) or ouabain (10 microM; block Na+-K+-ATPase) or after elevating external K+ concentration ([K+]) to 36 mM (to block K+ currents); curve fitting was performed to quantitate the rates of rise/fall and frequency under these conditions. Slow waves were markedly slowed, and eventually abolished, by niflumic acid but were unaffected by ouabain or high [K+]. Electrically evoked slow waves were also blocked in similar fashion by niflumic acid. We conclude that the repolarization phase is mediated by Ca2+-dependent Cl- currents. This information, together with our earlier finding that the depolarizing phase is due to voltage-dependent Ca2+ current, suggests that slow waves in canine BSM involve alternating opening and closing of Ca2+ and Cl- channels.
Collapse
Affiliation(s)
- L J Janssen
- Asthma Research Group and Smooth Muscle Research Program, Department of Medicine, McMaster University, Hamilton, Ontario, Canada L8N 3Z5
| | | | | |
Collapse
|
21
|
Hazama H, Nakajima T, Asano M, Iwasawa K, Morita T, Igarashi K, Nagata T, Horiuchi T, Suzuki J, Soma M, Okuda Y. Omega-3 polyunsaturated fatty acids--modulation of voltage-dependent L-type Ca2+ current in guinea-pig tracheal smooth muscle cells. Eur J Pharmacol 1998; 355:257-66. [PMID: 9760040 DOI: 10.1016/s0014-2999(98)00484-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Omega-3 polyunsaturated fatty acids have been reported to be associated with favorable changes in the respiratory system. To determine one of the mechanisms for this effect, membrane currents were recorded in guinea-pig tracheal myocytes by using the whole-cell voltage clamp technique. Without EGTA in the patch pipette containing the Cs-internal solution, command voltage pulses positive to +0 mV from a holding potential of -60 mV elicited a voltage-dependent L-type Ca2+ current (I(Ca x L)) and a subsequent outward current. Upon repolarization, slowly decaying inward tail currents were recorded. The outward currents and the inward tail current were enhanced by methyl-1,4,-dihydro-2,6-dimethyl-3-nitro-4-(2-trigluromethylphenyl )-pyridine-5-carboxylate, and blocked by Cd2+ or nifedipine. Inclusion of EGTA (5 mM) in the patch pipette also abolished these currents, indicating that they were Ca2+-dependent. When [Cl-]o or [Cl-]i was changed, the reversal potential of these currents shifted, thus behaving like a Cl(-)-sensitive ion channel. 4,4'-Diisothiocyanatostilbene-2,2'-disulphonic acid. a Cl- channel blocker, inhibited the currents. The omega-3 polyunsaturated fatty acids eicosapentaenoic acid (3-30 microM) and docosahexaenoic acid (30 microM) suppressed I(Ca x L) and then inhibited I(Ca x Cl) in a reversible manner. Similar inhibitory effects of eicosapentaenoic acid on I(Ca x L) were observed with 5 mM EGTA in the patch pipette. Neurokinin A (1 microM) and caffeine (10 mM) also transiently activated I(Cl x Ca), probably due to Ca2+ release from Ca2+ storage sites. Pretreatment of the cells with eicosapentaenoic acid markedly suppressed the activation of I(Cl x Ca) by neurokinin A or caffeine. These results suggest that omega-3 polyunsaturated fatty acids inhibit voltage-dependent L-type Ca2+ currents and also Ca2+-activated Cl- currents in tracheal smooth muscle cells from the guinea-pig, which may play a role in modulation of tracheal smooth muscle tone.
Collapse
Affiliation(s)
- H Hazama
- Second Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Li L, Vapaatalo H, Vaali K, Paakkari I, Kankaanranta H. Flufenamic and tolfenamic acids and lemakalim relax guinea-pig isolated trachea by different mechanisms. Life Sci 1998; 62:PL 303-8. [PMID: 9600333 DOI: 10.1016/s0024-3205(98)00157-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The effects of K+ channel inhibitors on the relaxations induced by flufenamic and tolfenamic acids and lemakalim were examined in guinea-pig isolated trachea precontracted with prostaglandin F2alpha (PGF2alpha, 1 microM). Flufenamic and tolfenamic acids (0.1-33 microM) and lemakalim (0.01-33 microM) relaxed guinea-pig trachea in a concentration-dependent manner. Tetraethylammonium (0.5-2 mM), a nonspecific inhibitor of K+ channels, inhibited the relaxations induced by flufenamic and tolfenamic acids without affecting lemakalim-induced relaxation. Charybdotoxin (ChTX, 33-100 nM), an inhibitor of the large Ca2+-activated K+ channels (BK(Ca)), also inhibited the relaxations induced by flufenamic and tolfenamic acids without affecting lemakalim-induced relaxation. Glipizide (3.3-33 microM), an inhibitor of the ATP-sensitive K+ channels (K(ATP)) inhibited lemakalim-induced relaxation without affecting those induced by flufenamic and tolfenamic acids. Our results indicate that the relaxations of guinea-pig isolated trachea by flufenamic and tolfenamic acids are due to activation of BK(Ca). The relaxant mechanism of flufenamic and tolfenamic acids thus differs from that of lemakalim, an activator of K(ATP).
Collapse
Affiliation(s)
- L Li
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Helsinki, Finland
| | | | | | | | | |
Collapse
|
24
|
Li L, Vapaatalo H, Vaali K, Paakkari I, Kankaanranta H. Fenamates inhibit contraction of guinea-pig isolated bronchus in vitro independent of prostanoid synthesis inhibition. Life Sci 1998; 62:PL289-94. [PMID: 9585115 DOI: 10.1016/s0024-3205(98)00146-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inhibitory and relaxant effects of flufenamic and tolfenamic acids on guinea-pig isolated bronchus were compared with those of verapamil and indomethacin. Flufenamic and tolfenamic acids (each drug, 20 microM) and verapamil (1 microM) inhibited bronchial contraction induced by Ca2+, KCl or PGF2alpha whereas indomethacin (20 microM) had no inhibitory effect. Only verapamil, but not flufenamic and tolfenamic acids and indomethacin, inhibited methacholine-induced contraction. Flufenamic and tolfenamic acids and verapamil (each drug, 0.1-33 microM) relaxed the bronchus precontracted by KCl or PGF2alpha. In contrast, indomethacin (0.1-33 microM) did not relax KCl- or PGF2alpha-precontracted bronchus. Verapamil, but not flufenamic and tolfenamic acids and indomethacin, relaxed methacholine precontracted bronchus. In conclusion, fenamates inhibit Ca2+-, KCl- and PGF2alpha-induced contractions in guinea-pig isolated bronchus in a manner involving inhibition of Ca2+ influx but not inhibition of prostanoid synthesis.
Collapse
Affiliation(s)
- L Li
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Helsinki, Finland
| | | | | | | | | |
Collapse
|
25
|
Li L, Kankaanranta H, Vaali K, Paakkari I, Vapaatalo H. Econazole, miconazole and SK & F 96365 inhibit depolarization-induced and receptor-operated contraction of guinea-pig isolated trachea in vitro. Eur J Pharmacol 1997; 331:221-5. [PMID: 9274983 DOI: 10.1016/s0014-2999(97)01068-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Econazole, miconazole, SK & F 96365 and nifedipine inhibited Ca2+- and depolarization-induced and receptor-operated contraction of guinea-pig isolated trachea. Econazole, miconazole and SK & F 96365 inhibited histamine- and methacholine-induced tracheal contraction more than nifedipine. Nifedipine was more potent in inhibiting KCl-induced contraction. Nifedipine, salbutamol and theophylline, but not econazole, miconazole or SK & F 96365, relaxed KCl, histamine-, and methacholine-precontracted trachea. It appears that in the guinea-pig tracheal smooth muscle, econazole, miconazole and SK & F 96365 behave differently from nifedipine, theophylline and salbutamol. Econazole, miconazole and SK & F 96365 are thus introduced as novel antagonists of receptor-operated airway smooth muscle contraction.
Collapse
Affiliation(s)
- L Li
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Helsinki, Finland
| | | | | | | | | |
Collapse
|
26
|
Bauer V, Oike M, Tanaka H, Inoue R, Ito Y. Hydrogen peroxide induced responses of cat tracheal smooth muscle cells. Br J Pharmacol 1997; 121:867-74. [PMID: 9222542 PMCID: PMC1564763 DOI: 10.1038/sj.bjp.0701202] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
1. The effects of hydrogen peroxide (H2O2) (10(-6)-10(-3) M) on membrane potential, membrane currents, intracellular calcium concentration, resting muscle tone and contractions elicited by electrical field stimulation (EFS) and carbachol were examined in cat tracheal strips and isolated smooth muscle cells. 2. H2O2 (10(-4) and 10(-5) M) enhanced the amplitude of contractions and excitatory junction potentials (e.j.p.) evoked by EFS without changing muscle tone and resting membrane potential of the tracheal smooth muscle, and enhanced the contraction induced by carbachol (10(-3) M). At an increased concentration (10(-3) M), H2O2 elevated resting muscle tone and marginally hyperpolarized the membrane in the majority of the cells. 3. In 51 out of 56 cells examined, H2O2 (10(-6)-10(-3) M) elicited an outward current at a holding potential of -40 mV and enhanced the frequency of the spontaneous transient outward current (STOC). In 20 cells the outward current was preceded by a small inward current. In the other cells, H2O2 elicited only an inward current or did not affect the background current. 4. In Ca2+ free solution the action of H2O2 on the resting muscle tone, STOCs, background current and on the current induced by ramp depolarization was significantly reduced. 5. H2O2 (10(-4) M) increased the intracellular ionized calcium concentration both in the absence and presence of external Ca2+. However, the effect developed faster and was of a higher amplitude in the presence of external Ca2+. 6. These results suggest that H2O2 increases intracellular Ca2+, with a subsequent augmentation of stimulation-evoked contractions, and enhances Ca2+ and voltage-sensitive potassium conductance.
Collapse
Affiliation(s)
- V Bauer
- Department of Pharmacology Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
27
|
Janssen LJ. T-type and L-type Ca2+ currents in canine bronchial smooth muscle: characterization and physiological roles. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 272:C1757-65. [PMID: 9227402 DOI: 10.1152/ajpcell.1997.272.6.c1757] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We examined the voltage-dependent Ca2+ currents in freshly dissociated smooth muscle cells obtained from canine bronchi (3rd to 5th order). When cells were depolarized from -40 mV, we observed an inward current that 1) exhibited threshold and peak activation at approximately -35 mV and +10 mV, respectively; 2) inactivated slowly with half-inactivation at -20 mV; 3) deactivated rapidly (time constant < 1 ms) upon repolarization; and 4) was abolished by nifedipine and suppressed by cholinergic agonist. These characteristics are typical of L-type Ca2+ current. During depolarization from -70 or -80 mV, however, many cells exhibited a second inward current superimposed on the L-type Ca2+ current. Activation of this other current was first noted at -60 mV, was maximal at approximately -20 mV, and was very rapid (reaching a peak within 10 ms). Inactivation of the other current was also rapid (time constant approximately 3 ms) and half-maximal at approximately -70 mV. There was a persistent "window" current over the physiologically relevant range of potentials (i.e., -60 to -30 mV). This current was also sensitive to nifedipine (although less so than the L-type current) and to Ni2+, but not to cholinergic agonist. Finally, the tail currents evoked upon repolarization to the holding potential decayed approximately 10 times more slowly than did L-type tail currents. These characteristics are all typical of T-type Ca2+ current. We conclude that there is a prominent T-type Ca2+ current in canine bronchial smooth muscle; this current may play a central role in excitation-contraction coupling, in refilling of the internal Ca2+ pool, and in electrical slow waves. Because airflow resistance is determined primarily by the smaller airways and not the trachea, these findings may have important implications with respect to airway physiology and the mechanisms underlying airway hyperreactivity and asthma.
Collapse
Affiliation(s)
- L J Janssen
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
28
|
Abstract
Spontaneous transient outward currents (STOCs) lasting about 100 ms occur in single smooth muscle cells and represent the simultaneous opening of up to a hundred calcium-activated potassium (BK) channels. The recent observation of brief focal releases of sarcoplasmic reticulum (SR) calcium ('sparks') in smooth muscle cells has provided support for the original suggestion that STOCs arise due to the spontaneous releases of calcium from the SR close to the sarcolemma. However, it is possible that such releases occur in a region of close apposition of SR membrane and sarcolemma about 0.1 microns wide ('junctional space') in which case they would be detectable by endogenous calcium-sensitive molecules such as BK channels but, using present confocal microscopy technique, not by calcium-indicator dyes introduced into the cell; should calcium escape from the junctional space then it may be visualised as 'sparks' by the fluorescent emission from calcium-indicator dyes using confocal microscopy. Some STOCs seem too large to represent the effect of a single 'spark' and some form of calcium-induced calcium release or 'macrospark' may be involved in their generation. Depletion of calcium stores by caffeine, ryanodine, or by activation of receptors linked to the phospholipase C/inositol trisphosphate system abolishes STOCs. However, low concentrations of caffeine or inositol trisphosphate accelerate STOC discharge by an unknown mechanism and often decrease STOC size presumably by depleting store calcium; similar effects are produced by agents such as cyclopiazonic acid and thapsigargin which inhibit calcium storage mechanisms (largely the SR calcium pump).
Collapse
Affiliation(s)
- T B Bolton
- Department of Pharmacology and Clinical Pharmacology, St George's Hospital Medical School, London, UK.
| | | |
Collapse
|
29
|
Henmi S, Imaizumi Y, Muraki K, Watanabe M. Time course of Ca(2+)-dependent K+ and Cl- currents in single smooth muscle cells of guinea-pig trachea. Eur J Pharmacol 1996; 306:227-36. [PMID: 8813636 DOI: 10.1016/0014-2999(96)00193-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The time course of two types of Ca(2+)-dependent currents were compared in single smooth muscle cells freshly isolated from guinea-pig trachea. When the pipette solution contained mainly 140 mM KCl, depolarization from -60 mV to 0 mV evoked an initial inward current followed by an outward current which consisted of transient (I(to)) and sustained components. In addition, a long-lasting inward tail current (Itail) was occasionally observed after the repolarization to -60 mV. Although I(to) often occurred repetitively during depolarization, the first I(to) reached the peak of approximately 50 ms after the start of depolarization and had the largest amplitude in most cells examined. The amplitude of Itail increased with the increase in depolarization period up to about 500 ms. Pharmacological analyses indicate that I(to) and Itail are Ca(2+)-dependent K+ and Cl- currents (IK-Ca and ICl-Ca), respectively, and suggest that not only Ca(2+)-influx through Ca2+ channels but also subsequent Ca2+ release from stores contributes to activate these currents. Spontaneous transient outward and inward currents, IK-Ca and ICl-Ca, respectively, were simultaneously recorded at -40 mV. In over 80% of the spontaneous current events, outward and inward currents coupled one to one and always occurred in this order. Puff-application of 10 mM caffeine also induced IK-Ca and ICl-Ca in this order at -40 mV. When caffeine was applied twice with various intervals, the current amplitude in the second application depended upon the period of the interval. The recovery of ICl-Ca during the interval was faster than that of IK-Ca. The results indicate that the activation and decay time courses of ICl-Ca are slower but its recovery is faster than those of IK-Ca.
Collapse
Affiliation(s)
- S Henmi
- Department of Chemical Pharmacology of Faculty of Pharmaceutical Sciences, Nagoya City University, Japan
| | | | | | | |
Collapse
|
30
|
Hazama H, Nakajima T, Hamada E, Omata M, Kurachi Y. Neurokinin A and Ca2+ current induce Ca(2+)-activated Cl(-) currents in guinea-pig tracheal myocytes. J Physiol 1996; 492 ( Pt 2):377-93. [PMID: 9019536 PMCID: PMC1158834 DOI: 10.1113/jphysiol.1996.sp021315] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
1. Membrane currents were recorded by a patch clamp technique in guinea-pig tracheal myocytes, using the whole cell mode with Cs(+) internal solution. 2. Both neurokinin A (NKA, 1 mu M) and caffeine (10 mM) evoked Ca(2+)-activated Cl- currents (I[Cl(Ca)]) transiently. In Ca(2+)-free bathing solution, the first application of NKA or caffeine elicited I[Cl(Ca)] but the second application of these substances failed to activate it. In addition, pretreatment with ryanodine in the presence of caffeine abolished the response to both NKA and caffeine whilst heparin (200 mu g ml(-1)) only blocked the NKA-induced response. I[Cl(Ca)] was also elicited by inositol 1,4,5-trisphosphate (IP(3)). 3. Command voltage pulses positive to 0 mV from a holding potential of -60 mV activated the voltage-dependent L-type Ca2+ current (I(Ca,L)) and late outward current. Upon repolarization to the holding potential, slowly decaying inward tail currents were recorded. The outward current during the depolarizing pulses and the inward tail current were enhanced by Bay K 8644, but completely blocked by Cd2+ or nifedipine. Replacement of external Ca2+ with Ba2+, removal of Ca2+ from the bath solution, or inclusion of EGTA (5 mM) in the patch pipette, also led to abolition of these currents, indicating that they were Ca2+ dependent, and that Ca2+ influx due to I(Ca,L) activated the currents. 4. When [Cl(-)](O) or [Cl(-)](i) was changed, the reversal potential (E(rev)) of the Ca2+-activated currents shifted, thus behaving like a Cl(-)-selective ion channel as predicted by the Nernst equation. DIDS (1 mM) completely abolished the currents, also suggesting that they were I[Cl(Ca)]. 5. NKA (1 mu M) and caffeine (30 mM) transiently activated I[Cl(Ca)], and after that both agents markedly reduced I[Cl(Ca)] induced by I(Ca,L). This is probably due to sarcoplasmic reticulum (SR) Ca2+ release induced by NKA or caffeine, followed by inhibition of the Ca(2+)-induced Ca2+ release from the SR. 6. The present results indicate that I[Cl(Ca)] can be activated by SR Ca2+ release due to NKA or caffeine (through IP(3) or ryanodine receptors) as well as by Ca2+ influx due to I(Ca,L). It also suggests that activation of I[Cl(Ca)] by NKA may be mediated by the production of IP(3), which releases Ca2+ from the SR.
Collapse
Affiliation(s)
- H Hazama
- The Second Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
31
|
Fleischmann BK, Wang YX, Pring M, Kotlikoff MI. Voltage-dependent calcium currents and cytosolic calcium in equine airway myocytes. J Physiol 1996; 492 ( Pt 2):347-58. [PMID: 9019534 PMCID: PMC1158832 DOI: 10.1113/jphysiol.1996.sp021313] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
1. The relationship between voltage-dependent calcium channel current (I(Ca)) and cytosolic free calcium concentration ([Ca2+]i) was studied in fura-2 AM-loaded equine tracheal myocytes at 35 degrees C and 1.8 mM Ca2+ using the nystatin patch clamp method. The average cytosolic calcium buffering constant was 77 +/- 3 (n = 14), and the endogenous calcium buffering constant component is likely to be between 15 and 50. 2. I(Ca) did not evoke significant calcium-induced calcium release (CICR) since (i)[Ca2+]i scaled with the integrated I(Ca) over the full voltage range of evoked calcium currents, (ii) increases in [Ca2+]i associated with I(Ca) were consistent with cytoplasmic buffering of calcium ions entering through voltage-dependent calcium channels (VDCCs) only, (iii) there was a fixed instantaneous relationship between transmembrane calcium flux (J(Ca)) and the change in cytosolic free calcium concentration (delta [Ca2+]i) during I(Ca), (iv) caffeine (8 mM) triggered 8-fold higher calcium transients than I(Ca), and (v) I(Ca) evoked following release of intracellular calcium by caffeine resulted in an equivalent delta[Ca2+]i-J(Ca) relationship. 3. The time constant (T) for the decay in [Ca2+]i was 8.6 +/- 1.5 s (n = 8) for single steps and 8.6 +/- 1.1 s (n = 13) following multiple steps that increased [Ca2+]i to much higher levels. Following application of caffeine (8 mM), however, [Ca2+]i decay was enhanced (T = 2.0 +/- 0.2 s, n = 3). The rate of [Ca2+]i decay was not voltage dependent, was not decreased in the absence of extracellular Na+ ions, and no pump current was detected. 4. We conclude that under near physiological conditions, neither CICR nor Na(+)-Ca2+ exchange play a substantial role in the regulation of I(Ca)-induced increases in [Ca2+]i, and that, even following release of intracellular calcium by caffeine, Na(+)-Ca2+ exchange does not play an appreciable role in the removal of calcium ions from the cytosol.
Collapse
Affiliation(s)
- B K Fleischmann
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104-6046, USA
| | | | | | | |
Collapse
|
32
|
Wade GR, Barbera J, Sims SM. Cholinergic inhibition of Ca2+ current in guinea-pig gastric and tracheal smooth muscle cells. J Physiol 1996; 491 ( Pt 2):307-19. [PMID: 8866856 PMCID: PMC1158727 DOI: 10.1113/jphysiol.1996.sp021217] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1. Cholinergic regulation of L-type Ca2+ channels was investigated in freshly dissociated guinea-pig gastric and tracheal smooth muscle cells. Acetylcholine (ACh, 50 microM) decreased Ca2+ channel current (ICa) by 37 +/- 3% (mean +/- S.E.M., 46 cells). 2. ACh reduced ICa at all voltages, with no shift in the current-voltage relationship. Effects of ACh were rapid (within 5 s) and repeatable, with multiple applications reproducibly inhibiting ICa in the continued presence of extracellular Ca2+ and in the presence of protein kinase C inhibitors. 3. The involvement of Ca2+ stores in this inhibition was investigated using Ca(2+)-free solution or cyclopiazonic acid (CPA) to deplete the stores. ACh initially inhibited ICa in the Ca(2+)-free solution (Na+ as charge carrier, 53 +/- 4% decrease, 18 cells) with subsequent responses significantly attenuated (n = 9). CPA (1 microM) reduced, then abolished, the effects of ACh on ICa (n = 5). 4. When studied in cell-attached patches (Ba2+ as charge carrier), ACh reduced Ca2+ channel open probability in twenty-two of thirty-six cells, consistent with the involvement of a diffusible cytosolic messenger. 5. ACh also inhibited ICa in tracheal muscle cells (reduction of 38 +/- 6% in 1 mM Ca2+, 4 cells; 77 +/- 3% in Ca(2+)-free solution, 7 cells). Furthermore, in cells where ACh elicited oscillating Ca(2+)-activated Cl- current, oscillatory inhibition of ICa was also observed (3 cells). 6. In summary, ACh causes rapid and reversible inhibition of ICa in gastric and tracheal muscles. Ca2+ stores were required to initiate this effect, with the rapid onset and oscillatory inhibition consistent with Ca2+ inhibition of the channel. Suppression of ICa would reduce Ca2+ entry during cholinergic excitation.
Collapse
Affiliation(s)
- G R Wade
- Department of Physiology, University of Western Ontario, London, Canada
| | | | | |
Collapse
|
33
|
Sadraei H, Beech DJ. Ionic currents and inhibitory effects of glibenclamide in seminal vesicle smooth muscle cells. Br J Pharmacol 1995; 115:1447-54. [PMID: 8564204 PMCID: PMC1908885 DOI: 10.1111/j.1476-5381.1995.tb16636.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
1. Whole-cell voltage-clamp recordings were made from smooth muscle cells isolated from guinea-pig seminal vesicle. 2. When the recording pipette solution contained 130 mM KCl and a low concentration of EGTA (0.2 mM), a dominant outward current was elicited by depolarization to positive of -30 mV from a holding potential of -50 mV. The current was non-inactivating, stimulated by intracellular Ca2+ and blocked by bath-applied 1 mM tetraethylammonium but not 1 mM 3,4 diaminopyridine. 3. If 10 mM EGTA was added to the KCl pipette solution and the holding potential was -50 mV, or more negative, the major current elicited by depolarization to positive of -30 mV was an A-type K(+)-current. This current inactivated rapidly (within 100 ms) and was blocked by bath-applied 1 mM 3,4-diaminopyridine but not 10 mM tetraethylammonium. 4. An inward voltage-gated Ca channel current was observed on depolarization to positive of -30 mV with 1.5 mM Ca2+ or 10 mM Ba2+ in the bath solution and when Ca+ replaced K+ in the pipette. The Ba(2+)-current was shown to be abolished by bath-applied 100 microM Cd2+ and inhibited by 90% by 1 microM nifedipine, and thus appeared to be carried by L-type Ca channels. 5. High concentrations of glibenclamide (10-500 microM) inhibited A-type K(+)-current, Ba(2+)-current and contraction of the whole tissue induced by noradrenaline or electrical field stimulation. 6. From these data we suggest that seminal vesicle smooth muscle cells express Ca2+ -dependent K channels, A-type K channels and L-type Ca channels which are inhibited by tetraethylammonium,3,4-diaminopyridine and nifedipine, respectively. In addition, an unexpected relaxant effect of high concentrations of glibenclamide may be explained by inhibition of the Ca channels.
Collapse
Affiliation(s)
- H Sadraei
- Department of Pharmacology, University of Leeds
| | | |
Collapse
|
34
|
Nakajima T, Hazama H, Hamada E, Omata M, Kurachi Y. Ionic basis of neurokinin-A-induced depolarization in single smooth muscle cells isolated from guinea-pig trachea. Pflugers Arch 1995; 430:552-62. [PMID: 7491282 DOI: 10.1007/bf00373892] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Neurokinin A (NKA) caused single tracheal smooth muscle cells (TSMCs) to contract. The effects of NKA on the electrical activity of guinea-pig TSMCs were examined using the tight-seal whole-cell patch-clamp technique. Under current-clamp conditions at rest, the membrane potential of TSMCs spontaneously oscillated at about -40 mV and NKA rapidly depolarized the membrane potential to nearly 0 mV, which then gradually repolarized to about -20 mV in the presence of NKA. The oscillations in potential disappeared transiently during the rapid phase of depolarization in response to NKA and reappeared during the sustained phase of depolarization. Under voltage-clamp conditions, NKA evoked an inward current which faded quickly. Subsequently, the cell conductance in the presence of NKA at potentials greater than -40 mV decreased gradually. The reversal potential of the NKA-induced inward current was about 0 mV, and shifted with changes in the Cl- equilibrium potential. The Cl- current was not elicited by NKA when using a pipette solution containing 10 mM ethylenebis(oxonitrilo)tetraacetic acid (EGTA). During the sustained phase, K+ currents evoked by depolarizing voltage steps were inhibited by NKA. The present results indicate that NKA causes rapid and sustained depolarization of TSMCs by two distinct mechanisms: (1) initial transient activation of the Ca(2+)-dependent Cl- current, and (2) sustained inhibition of K+ currents.
Collapse
Affiliation(s)
- T Nakajima
- Second Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- A J Knox
- Respiratory Medicine Unit, City Hospital, Nottingham, UK
| | | |
Collapse
|
36
|
Janssen LJ, Sims SM. Ca(2+)-dependent Cl- current in canine tracheal smooth muscle cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1995; 269:C163-9. [PMID: 7543242 DOI: 10.1152/ajpcell.1995.269.1.c163] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Our goal was to investigate the role of Ca2+ entry in regulating Cl- current (ICl) in smooth muscle cells from canine trachealis. When studies were done using the perforated patch configuration, depolarization elicited a dihydropyridine-sensitive Ca2+ current (ICa), followed in many cells by a sustained current. This sustained current reversed direction close to the Cl- equilibrium potential, consistent with its representing ICl. The ICl was also apparent as slowly deactivating tail currents seen upon repolarization to negative potentials. The Cl- channel blocker niflumic acid abolished both the sustained and tail currents, without affecting ICa. Several observations indicated that the ICl was dependent on Ca2+ entry. ICl was increased in magnitude when Ca2+ influx was augmented [by prolonging the depolarization or using BAY K 8644 or acetylcholine (ACh)] and decreased in magnitude when Ca2+ influx was reduced (using nifedipine). Based on these findings, we conclude that depolarization causes Ca2+ entry, with resultant elevation of cytosolic free Ca2+ concentration leading to activation of ICl (ICl(Ca)). We investigated whether Ca(2+)-induced Ca2+ release from the sarcoplasmic reticulum was involved in activation of ICl(Ca), by depleting intracellular stores of Ca2+ using cyclopiazonic acid to block the sarcoplasmic Ca(2+)-adenosinetriphosphatase and repeated stimulation with ACh. In such Ca(2+)-depleted cells, depolarization-mediated Ca2+ entry continued to activate ICl(Ca), suggesting that Ca(2+)-induced Ca2+ release was not required for its activation. We conclude that Ca2+ entry can activate Cl- channels in tracheal smooth muscle. This represents a positive-feedback system, which would promote excitation and contraction of airway muscle.
Collapse
Affiliation(s)
- L J Janssen
- Department of Physiology, University of Western Ontario, London, Canada
| | | |
Collapse
|
37
|
Lo YC, Wu SN, Wu JR, Chen IJ. Effect of capsaicin on membrane currents in cultured vascular smooth muscle cells of rat aorta. Eur J Pharmacol 1995; 292:321-8. [PMID: 7540981 DOI: 10.1016/0926-6917(95)90039-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The application of capsaicin (1 microM) produced a minor relaxant effect in endothelium-denuded rat aortae. However, capsaicin caused a greater relaxation of blood vessels precontracted with high K+ or phenylephrine. The effects of capsaicin on the ionic currents were also examined in A7r5 vascular smooth muscle cells. The tight-seal whole-cell voltage clamp technique was used. Capsaicin inhibited the Ba2+ inward current (IBa) through the voltage-dependent L-type Ca2+ channel in a concentration-dependent fashion, whereas calcitonin gene-related peptide and phenylephrine produced a minor increase in IBa. Capsaicin did not alter the overall shape of current-voltage relationship of IBa. However, capsaicin (3 microM) shifted the quasi-steady-state inactivation curve of IBa to more negative membrane potential by about 5 mV. These effects of capsaicin on IBa were reversible. In addition, capsaicin had inhibitory effects on voltage dependent K+ currents. These results suggest that inhibition of the voltage-dependent L-type Ca2+ channel is involved in the capsaicin-induced relaxation of the vascular smooth muscle, whereas capsaicin-induced inhibition of voltage-dependent K+ channels might produce an increase in cell excitability.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Capsaicin/pharmacology
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cells, Cultured
- Electrophysiology
- Endothelium, Vascular/physiology
- Ion Channels/drug effects
- Ion Channels/metabolism
- Muscle Contraction/drug effects
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Patch-Clamp Techniques
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Y C Lo
- Department of Pharmacology, Kaohsiung Medical College, Taiwan
| | | | | | | |
Collapse
|
38
|
Hazama H, Nakajima T, Hisada T, Hamada E, Omata M, Kurachi Y. Effects of azelastine on membrane currents in tracheal smooth muscle cells isolated from the guinea-pig. Eur J Pharmacol 1994; 259:143-50. [PMID: 7957608 DOI: 10.1016/0014-2999(94)90503-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Azelastine [4-(p-chlorobenzyl)-2-(hexahydro-1-methyl -1H-azepin-4-yl)-1-(2H)-phthalazinone hydrochloride], an anti-allergic agent, inhibited the high K(+)-induced contraction in tracheal smooth muscle cells isolated from the guinea-pig. In order to investigate the ionic mechanisms, we examined the effects of azelastine on membrane currents, using the tight-seal whole cell voltage clamp technique. Azelastine (1-100 microM) caused an inhibition of the Ba2+ inward current (IBa) through the voltage-dependent L-type Ca2+ channel in a concentration-dependent manner. The inhibitory effect of azelastine on IBa was fully reversible. The IC50 value for azelastine-induced inhibition of IBa was approximately 8 microM, and 100 microM azelastine completely suppressed IBa. Azelastine exerted mainly a tonic block of IBa but did not show use dependence. Azelastine (10 microM) shifted the quasi-steady-state inactivation curve of IBa to more negative membrane potentials by approximately -20 mV, suggesting that the inhibitory effect of azelastine on IBa was voltage-dependent. In addition, azelastine produced inhibitory actions on other membrane currents (i.e. the voltage-dependent transient outward K+ current and the Ca(2+)-activated oscillatory K+ current) at doses higher than 10 microM. These results suggest that azelastine inhibits the voltage-dependent L-type Ca2+ current in single tracheal smooth muscle cells, which may contribute to the anti-allergic actions of azelastine in airways.
Collapse
Affiliation(s)
- H Hazama
- Second Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Janssen LJ, Sims SM. Emptying and refilling of Ca2+ store in tracheal myocytes as indicated by ACh-evoked currents and contraction. THE AMERICAN JOURNAL OF PHYSIOLOGY 1993; 265:C877-86. [PMID: 8238312 DOI: 10.1152/ajpcell.1993.265.4.c877] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Membrane currents and contractions evoked by acetylcholine (ACh) in freshly dissociated canine tracheal myocytes were investigated using the nystatin perforated-patch recording technique. In cells held at -60 mV in the presence of nifedipine, ACh evoked inward current (IACh) and contraction. Caffeine mimicked the effects of ACh. IACh and contractions could be evoked 3-4 min after removing external Ca2+ but were abolished by prolonged exposure to Ca(2+)-free media. Both responses were restored within minutes of reintroduction of Ca2+, even though the cells were held at -60 mV in the presence of nifedipine. IACh and ACh-evoked contractions were also reversibly abolished by continued exposure to caffeine. Cyclopiazonic acid (CPA), a blocker of the sarcoplasmic reticulum (SR) Ca(2+)-ATPase, reduced IACh by > 95% within 15 min but had little or no effect on the contractile responses evoked by ACh. IACh was restored after washout of CPA even though cells were held at -60 mV. After depleting the Ca2+ store with the use of CPA, depolarization of the membrane to +10 mV immediately before application of ACh led to a partial restoration of IACh. This restorative effect of depolarization was potentiated by Bay K 8644 and antagonized by nifedipine. In conclusion, IACh and contractions in canine tracheal myocytes are mediated by Ca2+ released from an internal store that can be depleted by prolonged removal of extracellular Ca2+, prolonged exposure to caffeine, or by blockade of the SR Ca(2+)-ATPase. At least two Ca2+ influx pathways appear to contribute to refilling of the internal store: one pathway that is not activated by depolarization or ACh and a second involving dihydropyridine-sensitive voltage-activated Ca2+ channels that may be in direct contact with the SR (i.e., conduct extracellular Ca2+ directly into the SR, bypassing the cytosol).
Collapse
Affiliation(s)
- L J Janssen
- Department of Physiology, University of Western Ontario, London, Canada
| | | |
Collapse
|
40
|
Fleischmann BK, Washabau RJ, Kotlikoff MI. Control of resting membrane potential by delayed rectifier potassium currents in ferret airway smooth muscle cells. J Physiol 1993; 469:625-38. [PMID: 8271220 PMCID: PMC1143891 DOI: 10.1113/jphysiol.1993.sp019834] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
1. In order to determine the physiological role of specific potassium currents in airway smooth muscle, potassium currents were measured in freshly dissociated ferret trachealis cells using the nystatin-permeabilized, whole-cell method, at 35 degrees C. 2. The magnitude of the outward currents was markedly increased as bath temperature was increased from 22 to 35 degrees C. This increase was primarily due to the increase in maximum potassium conductance (gK,max), although there was also a small leftward shift in the relationship between gK and voltage at higher temperatures. The maximum conductance and the kinetics of current activation and inactivation were also temperature dependent. At 35 degrees C, gating of the current was steeply voltage dependent between -40 and 0 mV. Current activation was well fitted by fourth-order kinetics; the mean time constants of activation (30 mV clamp step) were 1.09 +/- 0.17 and 1.96 +/- 0.27 ms at 35 and 22 degrees C, respectively. 3. Outward currents using the nystatin method were qualitatively similar to delayed rectifier currents recorded in dialysed cells with high calcium buffering capacity solutions. 4-Aminopyridine (4-AP; 2 mM), a specific blocker of delayed rectifier potassium channels in this tissue, inhibited over 80% of the outward current evoked by voltage-clamp steps to between -10 and +20 mV (n = 6). Less than 5% of the outward current was blocked over the same voltage range by charybdotoxin (100 nM; n = 15), a specific antagonist of large-conductance, calcium-activated potassium channels in this tissue. 4. The degree to which delayed rectifier and calcium-activated potassium conductances control resting membrane potential was examined in current-clamp experiments. The resting membrane potential of current clamped cells was -33.6 +/- 1.0 mV (n = 62). Application of 4-AP (2 mM) resulted in a 14.4 +/- 1.0 mV depolarization (n = 8) and an increase in input resistance. Charybdotoxin (100 nM) had no effect on resting membrane potential (n = 6). 5. Force measurements were made in isolated strips of trachealis muscle to determine the effect of pharmacological blockade of individual potassium conductances on resting tone. In the presence of tetrodotoxin (1 microM) and atropine (1 microM), 4-AP increased baseline tension in a dose-dependent manner, with an EC50 of 1.8 mM (n = 13); application of 5 mM 4-AP increased tone to 86.8 +/- 8.1% of that produced by 1 microM methacholine, and this tone was almost completely inhibited by nifedipine (1 microM).(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- B K Fleischmann
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104-6046
| | | | | |
Collapse
|
41
|
Abstract
Potassium channels are an important determinant of smooth muscle excitability and force generation. Two potassium channels have been fully described in airway smooth muscle: large conductance, calcium-activated potassium channels and voltage-dependent delayed rectifier channels. This article will review the biophysics and pharmacology of these channels and discuss what is currently known with respect to their regulation and physiological significance.
Collapse
Affiliation(s)
- M I Kotlikoff
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104-6046
| |
Collapse
|
42
|
Zholos AV, Baidan LV, Shuba MF. Some properties of Ca(2+)-induced Ca2+ release mechanism in single visceral smooth muscle cell of the guinea-pig. J Physiol 1992; 457:1-25. [PMID: 1338455 PMCID: PMC1175715 DOI: 10.1113/jphysiol.1992.sp019362] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
1. Late transient outward Ca(2+)-dependent K+ current (ILTO) correlated with Ca(2+)-induced Ca2+ release mechanism was studied in relation to the calcium inward current (ICa) in single isolated smooth muscle cells of the guinea-pig ileum using the whole-cell patch-clamp technique. 2. The voltage dependencies of peak ICa and ILTO were both bell shaped. However, the I-V curve of the outward current was shifted toward more positive potentials by about 60 mV in comparison to that for ICa. 3. Reduction in the external Ca2+ concentration resulted in a decrease of peak amplitude of both ICa and ILTO. However, caffeine-induced outward current was also decreased abruptly suggesting a rapid loss of stored Ca2+ upon lowering the external Ca2+ concentration. 4. Investigation of the relation of ILTO to partially inactivated ICa showed that inactivation of ICa by approximately 65, 80 or 84% of control (produced by prepulse to -20 mV for 2 s, shifting the holding potential to -20 mV for 30 s or by the ramp voltage command from -50 to +10 mV, respectively) was without detectable effect on the ILTO generation. 5. Bath application of the Ca2+ antagonist nifedipine (300 nM) inhibited ICa by 81% without affecting ILTO peak amplitude (92.0 +/- 5.6% of control in six cells). The mean concentration-response curve for ICa inhibition was sigmoidal with the apparent dissociation constant of 86.9 nM, whereas that for the ILTO had a characteristic sharp transition indicating a definite threshold of Ca2+ influx for ILTO generation. 6. Application of Ca(2+)-free external solution during 500 ms of the time when ICa peaked inhibited the current by about 76% whereas the ILTO during such an intervention remained virtually unchanged. 7. In double-pulse experiments, with conditioning and test pulses to +10 mV from -50 mV and an interpulse interval of 600 ms, most of the cells (about 80%) showed larger outward current at the test pulse suggesting continued Ca2+ release triggered by Ca2+ influx during a short (50-200 ms) depolarizing prepulse. The outward current could also be evoked at large positive potentials (presumably near the calcium equilibrium potential) where it did not occur normally by a prepulse to +10 mV for 50 ms. The charge transferred by Ca2+ current necessary to activate Ca2+ release in most of the cells was estimated to be from 6 to 20 pC. 8. The data are interpreted to suggest that the Ca(2+)-induced Ca2+ release mechanism operates in single ileal cells in a regenerative manner.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- A V Zholos
- Department of Nerve-Muscle Physiology, A.A. Bogomoletz Institute of Physiology, Academy of Sciences of the Ukraine, Kiev
| | | | | |
Collapse
|
43
|
Smirnov SV, Zholos AV, Shuba MF. Potential-dependent inward currents in single isolated smooth muscle cells of the rat ileum. J Physiol 1992; 454:549-71. [PMID: 1335506 PMCID: PMC1175620 DOI: 10.1113/jphysiol.1992.sp019279] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
1. Calcium (ICa) and sodium (INa) currents were studied in single smooth muscle cells freshly isolated from both the newborn (1-3 days old) and adult rat ileum, using the patch-clamp technique (whole-cell configuration). 2. Under conditions when INa was blocked, two components of ICa, low-voltage activated or ICa,low and high-voltage activated or ICa,high, were observed in the newborn rat ileal cells. ICa,high and ICa,low have differing voltage ranges of activation and steady-state inactivation and time courses of recovery from inactivation. Potential dependence of ICa,low was much steeper and shifted toward negative membrane potential than that for ICa,high (slope factors and the potential of half-maximal inactivation were 13.6 and -60.6 and 8.8 and -49 mV for ICa,low and ICa,high, correspondingly). 3. Nifedipine at the high concentration of 30 microM exerted no effect on ICa,low and only slightly suppressed ICa,high, decreasing its peak to 0.81 +/- 0.04 (n = 7) at the holding potential of -80 mV and to 0.66 +/- 0.05 (n = 3) at -50 mV. ICa,high was suppressed significantly by Cd2+ ions, while ICa,low was more sensitive to Ni2+ ions. 4. Results presented here suggest that the properties of high-voltage-activated (HVA) Ca2+ channels in the rat small intestine are quite different to those described for L-type Ca2+ channels found in other smooth muscles. It is proposed that HVA Ca2+ channels are similar to N-type Ca2+ channels. 5. Comparison of Ca2+ currents in newborn and adult rat ileal cells showed that the contribution of ICa,low to the net Ca2+ current was negligible in adults, whereas the properties of HVA Ca2+ channels were similar in the neonatal and adult animals. 6. INa, studied in nominally Ca(2+)-free physiological salt solution, activated in the voltage range between -50 and -40 mV and reached its peak at -10 mV. INa was blocked in a dose-dependent manner by TTX with an apparent dissociation constant of 4.5 nM. 7. INa decay was monoexponential in the voltage range studied and its time constant decreased monotonically with membrane depolarization from 4.7 +/- 0.2 ms (n = 6) at -30 mV to 0.51 +/- 0.03 ms (n = 7) at 20 mV.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- S V Smirnov
- Department of Nerve-Muscle Physiology, A. A. Bogomoletz Institute of Physiology, Academy of Ukrainian Sciences, Kiev
| | | | | |
Collapse
|
44
|
Smirnov SV, Zholos AV, Shuba MF. A potential-dependent fast outward current in single smooth muscle cells isolated from the newborn rat ileum. J Physiol 1992; 454:573-89. [PMID: 1474503 PMCID: PMC1175621 DOI: 10.1113/jphysiol.1992.sp019280] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
1. Whole-cell outward currents have been studied in single smooth muscle cells isolated from newborn and adult rat ileum, using fire-polished glass micropipettes. 2. Two major outward currents, delayed (I(do)) and fast inactivating potential-dependent (I(fo)), have been observed in the newborn rat ileal cells. I(fo) is activated between -50 and -40 mV from the holding potential of -80 mV, whereas I(do) usually starts to activate at membrane potentials positive to -20 mV. Activation of I(do) was fast, its time-to-peak decreased from 10.8 +/- 0.9 ms (n = 5) at -30 mV to 4.5 +/- 0.7 ms (n = 4) at 20 mV. 3. I(fo) decay was monoexponential and its time constant did not depend on the membrane potential. Dependence of I(fo) inactivation on membrane voltage in normal physiological salt solutions (PSS) could be described by the Boltzmann equation with the following parameters: a half-inactivation potential, V0.5 = -70.8 mV and slope factor, k = 7.7 mV. 4. Recovery of I(fo) from inactivation was fitted by a single exponential and was potential dependent. The average time constant was 28.4 +/- 2.4 ms (n = 11) at -120 mV, 47.7 +/- 3.0 ms (n = 6) at -100 mV and 89.6 +/- 5.3 ms (n = 13) at -80 mV. 5. Removal of Ca2+ ions from the PSS (in the presence of 5 mM-Mg2+) increased I(fo) amplitude by about two times, and shifted its voltage dependence of inactivation towards negative membrane potentials by about 16 mV (V0.5 = -87.2 mV). Removal of Mg2+ from the PSS (in the presence of 2.5 mM-Ca2+) exerted no effects upon either inactivation dependence (V0.5 = -74.2 mV) or I(fo) amplitude. 6. I(do) and I(fo) had different sensitivities to K+ channel blockers. With 10 mM-external TEA+ I(do), was preferentially suppressed, while 5 mM-4-aminopyridine (4-AP) completely blocked I(fo). I(fo) was also partially blocked by a higher TEA+ concentration (30 mM), which suppressed I(fo) to 0.55 +/- 0.02 (n = 9). The blocking effect of 4-AP on I(fo) was potential, use and time dependent. 7. Ileal cells isolated from the adult rat demonstrated the presence of two populations of smooth muscle cells. One has an outward current which seems to be similar to that described in the newborn rat. However, in other cells spontaneous transient outward currents, well described in other single smooth muscle cells, but not found in newborn rat ileal cells, have been observed.
Collapse
Affiliation(s)
- S V Smirnov
- Department of Nerve-Muscle Physiology, A.A. Bogomoletz Institute of Physiology, Academy of Ukrainian Sciences, Kiev
| | | | | |
Collapse
|
45
|
McFadzean I, England S. Properties of the inactivating outward current in single smooth muscle cells isolated from the rat anococcygeus. Pflugers Arch 1992; 421:117-24. [PMID: 1382262 DOI: 10.1007/bf00374817] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The properties of the voltage- and time-dependent outward current in single smooth muscle cells isolated from the rat anococcygeus were studied. The outward current was activated by depolarizations to membrane potentials positive to -40 mV. Activation followed third order kinetics; at +20 mV, the time for the current to reach half its maximal amplitude was around 55 ms. The current inactivated with a time course that could best be described by a single exponential with a time constant around 1500 ms. The steady-state inactivation curve was voltage dependent over the range -110 to -30 mV, with a half-inactivation point of -67 mV. Recovery from inactivation followed an exponential time course with a time constant of around 770 ms at -90 mV. Deactivating tail current analysis revealed that a 10-fold change in the extracellular potassium ion concentration resulted in a 42 mV change in the reversal potential of the current. The current was blocked by 4-aminopyridine, tetraethylammonium, quinine and verapamil with IC50's--the concentrations producing 50% inhibition of the peak current--of 2 mM, 4 mM, 12 microM and 20 microM respectively. The current was not blocked by Toxin I (100 nM) or glibenclamide (10 microM). The current was still present in cells containing 5 mM EGTA; in these cells, replacing extracellular calcium with cadmium depressed the peak current by around 12%. This could be explained, at least in part, by a negative shift in the voltage dependence of inactivation.
Collapse
Affiliation(s)
- I McFadzean
- Division of Biomedical Sciences, King's College London, UK
| | | |
Collapse
|
46
|
Mironneau J, Yamamoto T, Sayet I, Arnaudeau S, Rakotoarisoa L, Mironneau C. Effect of dihydropyridines on calcium channels in isolated smooth muscle cells from rat vena cava. Br J Pharmacol 1992; 105:321-8. [PMID: 1373097 PMCID: PMC1908639 DOI: 10.1111/j.1476-5381.1992.tb14253.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
1. Whole-cell patch-clamp method was applied to single smooth muscle cells freshly isolated from the rat inferior vena cava. 2. Depolarizing pulses, applied from a holding potential of -90 mV, activated both Na+ and Ca2+ channels. The fast Na+ current was inhibited by nanomolar concentrations of tetrodotoxin (TTX). The slow Ba2+ current (measured in 5 mM Ba2+ solution) was inhibited by Cd2+ and modulated by dihydropyridine derivatives. When the cells were held at a holding potential of -80 mV, racemic Bay K 8644 increased the Ba2+ current (ED50 = 10 nM) while racemic isradipine inhibited the current (IC50 = 21 nM). 3. The voltage-dependency of isradipine blockade was assessed by determining the steady-state availability of the Ca2+ channels. From the shift of the inactivation curve in the presence of isradipine, we calculated a dissociation constant of 1.11 nM for inactivated Ca2+ channels. Scatchard plots of the specific binding of (+)-[3H]-isradipine obtained in intact strips incubated in 5.6 mM or 135 mM K+ solutions confirmed the voltage-dependency of isradipine binding. 4. Specific binding of (+)-[3H]-isradipine was completely displaced by unlabelled (+/-)-isradipine, with an IC50 of 15.1 nM. This value is similar to the IC50 for inhibition of the Ba2+ current (21 nM) in cells maintained at a holding potential of -80 mV. 5. Bay K 8644 had no effects on the Ba2+ current kinetics during a depolarizing test pulse. The steady-state inactivation-activation curves of Ba2+ current were not significantly shifted along the voltage axis.6. The present data suggest the existence of two distinct dihydropyridine binding sites which can be bound preferentially by agonist or antagonist derivatives.
Collapse
Affiliation(s)
- J Mironneau
- Laboratoire de Physiologie Cellulaire et Pharmacologie Moléculaire, INSERM CJF 88-13, Université de Bordeaux II, France
| | | | | | | | | | | |
Collapse
|
47
|
Yamazawa T, Iino M, Endo M. Presence of functionally different compartments of the Ca2+ store in single intestinal smooth muscle cells. FEBS Lett 1992; 301:181-4. [PMID: 1568477 DOI: 10.1016/0014-5793(92)81243-f] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Studies in smooth muscle bundles have shown the presence of functionally different compartments of Ca2+ store, one (S alpha) sensitive to both caffeine and inositol 1,4,5-trisphosphate (IP3), and the other (S beta) sensitive only to IP3. Ca2+ release in isolated single smooth muscle cells from guinea pig taenia caeci was studied to see if both compartments exist within a cell. Responses to caffeine and carbachol were consistently observed but were abolished after treatment with ryanodine, while intracellular application of IP3 induced Ca2+ release after the treatment, albeit smaller in size than control. Thus S alpha and S beta coexist in a single smooth muscle cell and agonist-induced Ca2+ release requires whole store to be loaded with Ca2+.
Collapse
Affiliation(s)
- T Yamazawa
- Department of Pharmacology, Faculty of Medicine, University of Tokyo, Japan
| | | | | |
Collapse
|
48
|
Boyle JP, Tomasic M, Kotlikoff MI. Delayed rectifier potassium channels in canine and porcine airway smooth muscle cells. J Physiol 1992; 447:329-50. [PMID: 1593449 PMCID: PMC1176039 DOI: 10.1113/jphysiol.1992.sp019005] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
1. In order to define the ion channels underlying the inactivating, calcium-insensitive current in airway smooth muscle cells, unitary potassium currents were recorded from canine and porcine trachealis cells, and compared with macroscopic currents. On-cell and inside-out single-channel currents were compared with whole-cell recordings made in dialysed cells. 2. Depolarizing voltage steps evoked outward unitary currents. In addition to a large conductance, calcium-activated potassium channel (KCa), a lower conductance potassium channel was identified. This channel has a conductance of 12.7 pS (on-cell; 1 mM-K+ in the pipette). 3. The lower conductance channel (Kdr) was not sensitive to cytosolic Ca2+ concentration and unitary current openings occurred following a delay after the voltage step. The time course of activation of the current composed of averaged single-channel events was very similar to that of the whole-cell, delayed rectifier potassium current (IdK), recorded under conditions of low intracellular calcium (Kotlikoff, 1990). 4. Kdr channels also inactivated with kinetics similar to those of the macroscopic current. Averaged single-channel records revealed a current that inactivated with kinetics that could be described by two exponentials (tau 1 = 0.14 s, tau 2 = 1.1 s; at 5 mV). These values corresponded well with previously determined values for time-dependent inactivation of IdK. Inactivation of Kdr channels was markedly voltage dependent, and was well fitted by a Boltzmann equation with V50 = -53 mV; this was similar to measurements of the macroscopic current, although the V50 value was shifted to more positive potentials in whole-cell measurements. When only the inactivating component of the macroscopic current was considered, the voltage dependence of inactivation of the single-channel current and macroscopic current were quite similar. 5. Single-channel kinetics indicated that Kdr channels occupy one open and two closed states. The mean open time was 1.7 ms. Inactivation results in a prominent increase in the long closed time, with little effect on the mean open time or short closed time. 6. The Kdr channel was not blocked by tetraethylammonium (TEA; 1 mM), charybdotoxin (ChTX; 100 nM) or glibenclamide (20 microM), but was blocked by 4-aminopyridine (4-AP; 1 mM). Similarly, 4-AP blocked the inactivating component of the macroscopic current, but a non-inactivating current remained. KCa currents were blocked by TEA (0.5-1 mM) and charybdotoxin (40 nM), but were insensitive to to 4-AP (1 mM) and glibenclamide (20 microM).(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- J P Boyle
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104-6046
| | | | | |
Collapse
|
49
|
Janssen LJ, Sims SM. Acetylcholine activates non-selective cation and chloride conductances in canine and guinea-pig tracheal myocytes. J Physiol 1992; 453:197-218. [PMID: 1281502 PMCID: PMC1175553 DOI: 10.1113/jphysiol.1992.sp019224] [Citation(s) in RCA: 139] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
1. Membrane currents activated by acetylcholine (ACh) were investigated in isolated canine and guinea-pig tracheal myocytes using the nystatin perforated patch configuration of whole-cell recording. ACh caused depolarization accompanied by a membrane conductance increase. 2. When cells were held under voltage clamp (holding potential, Vh = -60 mV), ACh elicited inward current (IACh) of up to 3900 pA, with a reversal potential (Erev) of approximately -20 mV. 3. Removal of extracellular Na+ (Na+o) reduced but did not eliminate IACh. IACh remaining in the absence of Na+ reversed direction close to the predicted equilibrium potential for Cl-. Erev shifted 32 +/- 4 mV per 10-fold change of [Cl-]i. Increasing external [K+] caused Erev to shift in the positive direction. These results suggest that ACh activated chloride and non-selective cation conductances. 4. In the absence of Na+o, the Cl- channel blockers SITS or niflumic acid reversibly antagonized IACh. 5. Caffeine and ryanodine elicited currents both in the presence and absence of Na+o; these currents had a reversal potential similar to that of IACh. Caffeine applied before ACh occluded the response to ACh. 6. We also observed two types of spontaneous membrane currents. Spontaneous transient outward currents (STOCs) may represent Ca(2+)-activated K+ currents. Spontaneous inward currents were also observed which were reduced in magnitude (but not eliminated) by removal of Na+o and reversed direction at approximately the Cl- equilibrium potential. The spontaneous inward currents and STOCs were coincident and were reversibly suppressed by ACh. 7. ACh elicited contractions of cells under voltage clamp at -60 mV, an effect also observed in the absence of extracellular Ca2+ or when IACh was reduced by omission of Na+o and exposure to Cl- channel blockers. The number of cells which did contract in response to ACh decreased, however, when the concentration of internal Cl- decreased. 8. All effects of ACh on contraction and membrane currents were antagonized by atropine. 9. We conclude that activation of muscarinic receptors in mammalian tracheal myocytes causes release of Ca2+ from intracellular stores and subsequent activation of Cl- and non-selective cation conductances. This is the first direct demonstration of these conductances in tracheal smooth muscle cells. Activation of these conductances does not appear to be required for contraction. However, regulation of cytosolic Cl- levels may be important for release and uptake of Ca2+ from internal stores.
Collapse
Affiliation(s)
- L J Janssen
- Department of Physiology, University of Western Ontario, London, Canada
| | | |
Collapse
|
50
|
Zholos AV, Baidan LV, Shuba MF. Properties of the late transient outward current in isolated intestinal smooth muscle cells of the guinea-pig. J Physiol 1991; 443:555-74. [PMID: 1726593 PMCID: PMC1179859 DOI: 10.1113/jphysiol.1991.sp018851] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
1. Whole-cell membrane currents in voltage-clamped single isolated cells of longitudinal smooth muscle of guinea-pig ileum were studied at room temperature using patch pipettes filled with either high-K+ solution or high-Cs+ solution, to suppress K+ outward current, and containing 0.3 mM-EGTA. 2. In the presence of high-K+ solution in the pipette, membrane depolarization from the holding potential of -50 mV evoked an initial inward calcium current (ICa) followed by a large initial transient outward current and a sustained outward current with spontaneous oscillations superimposed. Prolonged depolarization above -20 mV produced a late transient outward current which reached a maximum (up to several nanoamps at +10 mV) within approximately 1 s and lasted several seconds. 3. The late outward current (ILTO) was voltage dependent and reversed at the EK (potassium equilibrium potential) in cells exposed to high-K+ external solution. It was blocked by TEA+ (tetraethylammonium) or Ba2+ applied externally (calculated Kd (dissociation constant) values were 0.67 and 4.43 mM, respectively) or by high-Cs+ solution perfusing the cell. The removal of extracellular Ca2+, application of Ca2+ channel blockers (3 mM-Co2+, 0.2 mM-Cd2+ or 1 microM-nifedipine) or perfusion of 5 mM-EGTA inside the cell also abolished the current. Thus, the current seems to be a Ca(2+)-activated K+ current. 4. There is a great discrepancy between the time course of the ICa and that of the late ILTO, which suggests that Ca2+ release from intracellular storage sites may contribute to the generation of the ILTO. 5. Bath application of caffeine (10 mM) during the development of ILTO enhanced the current. However, in the presence of caffeine ILTO was inhibited. Moderate inhibition of ICa by caffeine was also observed. 6. Ryanodine (5 microM) applied to the bathing solution completely inhibited ILTO within 3.5 min; however, it had no or little effect on the ICa. 7. Ruthenium Red (10 microM) completely blocked the ILTO and slightly and more slowly inhibited the ICa. 8. Increasing Mg2+ concentration in the pipette solution from 1 to 6 mM abolished the ILTO. 9. It was concluded that the ILTO was activated mainly by Ca2+ released from the intracellular storage sites following Ca2+ entry, presumably by a Ca(2+)-induced Ca2+ release mechanism.
Collapse
Affiliation(s)
- A V Zholos
- Department of Nerve-Muscle Physiology, A. A. Bogomoletz Institute of Physiology, Academy of Sciences of the Ukrainian SSR, Kiev
| | | | | |
Collapse
|