51
|
Morales-Cano D, Moreno L, Barreira B, Pandolfi R, Chamorro V, Jimenez R, Villamor E, Duarte J, Perez-Vizcaino F, Cogolludo A. Kv7 channels critically determine coronary artery reactivity: left-right differences and down-regulation by hyperglycaemia. Cardiovasc Res 2015; 106:98-108. [PMID: 25616413 DOI: 10.1093/cvr/cvv020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIMS Voltage-gated potassium channels encoded by KCNQ genes (Kv7 channels) are emerging as important regulators of vascular tone. In this study, we analysed the contribution of Kv7 channels to the vasodilation induced by hypoxia and the cyclic AMP pathway in the coronary circulation. We also assessed their regional distribution and possible impairment by diabetes. METHODS AND RESULTS We examined the effects of Kv7 channel modulators on K+ currents and vascular reactivity in rat left and right coronary arteries (LCAs and RCAs, respectively). Currents from LCA were more sensitive to Kv7 channel inhibitors (XE991, linopirdine) and activators (flupirtine, retigabine) than those from RCA. Accordingly, LCAs were more sensitive than RCAs to the relaxation induced by Kv7 channel enhancers. Likewise, relaxation induced by the adenylyl cyclase activator forskolin and hypoxia, which were mediated through Kv7 channel activation, were greater in LCA than in RCA. KCNQ1 and KCNQ5 expression was markedly higher in LCA than in RCA. After incubation with high glucose (HG, 30 mmol/L), myocytes from LCA, but not from RCA, were more depolarized and showed reduced Kv7 currents. In HG-incubated LCA, the effects of Kv7 channel modulators and forskolin were diminished, and the expression of KCNQ1 and KCNQ5 was reduced. Finally, vascular responses induced by Kv7 channel modulators were impaired in LCA, but not in RCA, from type 1 diabetic rats. CONCLUSION Our results reveal that the high expression and function of Kv7 channels in the LCA and their down-regulation by diabetes critically determine the sensitivity to key regulators of coronary tone.
Collapse
Affiliation(s)
- Daniel Morales-Cano
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Rachele Pandolfi
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Virginia Chamorro
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Rosario Jimenez
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada 18071, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada 18071, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, Madrid 28040, Spain Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
52
|
Yoo HY, Park SJ, Kim HJ, Kim WK, Kim SJ. Integrative understanding of hypoxic pulmonary vasoconstriction using in vitro models: from ventilated/perfused lung to single arterial myocyte. Integr Med Res 2014; 3:180-188. [PMID: 28664095 PMCID: PMC5481745 DOI: 10.1016/j.imr.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 10/25/2022] Open
Abstract
Contractile response of a pulmonary artery (PA) to hypoxia (hypoxic pulmonary vasoconstriction; HPV) is a unique physiological reaction. HPV is beneficial for the optimal distribution of blood flow to differentially ventilated alveolar regions in the lung, thereby preventing systemic hypoxemia. Numerous in vitro studies have been conducted to elucidate the mechanisms underlying HPV. These studies indicate that PA smooth muscle cells (PASMCs) sense lowers the oxygen partial pressure (PO2) and contract under hypoxia. As for the PO2-sensing molecules, a variety of ion channels in PASMCs had been suggested. Nonetheless, the modulator(s) of the ion channels alone cannot mimic HPV in the experiments using PA segments and/or isolated organs. We compared the hypoxic responses of PASMCs, PAs, lung slices, and total lungs using a variety of methods (e.g., patch-clamp technique, isometric contraction measurement, video analysis of precision-cut lung slices, and PA pressure measurement in ventilated/perfused lungs). In this review, the relevant results are compared to provide a comprehensive understanding of HPV. Integration of the influences from surrounding tissues including blood cells as well as the hypoxic regulation of ion channels in PASMCs are indispensable for insights into HPV and other related clinical conditions.
Collapse
Affiliation(s)
- Hae Young Yoo
- Red Cross College of Nursing, Chung-Ang University, Seoul, Korea
| | - Su Jung Park
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Hae Jin Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Woo Kyung Kim
- Department of Internal Medicine and Channelopathy Research Institute (CRC), College of Medicine, Dongguk University, Goyang, Korea
| | - Sung Joon Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
53
|
Sedivy V, Joshi S, Ghaly Y, Mizera R, Zaloudikova M, Brennan S, Novotna J, Herget J, Gurney AM. Role of Kv7 channels in responses of the pulmonary circulation to hypoxia. Am J Physiol Lung Cell Mol Physiol 2014; 308:L48-57. [PMID: 25361569 PMCID: PMC4281702 DOI: 10.1152/ajplung.00362.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a beneficial mechanism that diverts blood from hypoxic alveoli to better ventilated areas of the lung, but breathing hypoxic air causes the pulmonary circulation to become hypertensive. Responses to airway hypoxia are associated with depolarization of smooth muscle cells in the pulmonary arteries and reduced activity of K+ channels. As Kv7 channels have been proposed to play a key role in regulating the smooth muscle membrane potential, we investigated their involvement in the development of HPV and hypoxia-induced pulmonary hypertension. Vascular effects of the selective Kv7 blocker, linopirdine, and Kv7 activator, flupirtine, were investigated in isolated, saline-perfused lungs from rats maintained for 3–5 days in an isobaric hypoxic chamber (FiO2 = 0.1) or room air. Linopirdine increased vascular resistance in lungs from normoxic, but not hypoxic rats. This effect was associated with reduced mRNA expression of the Kv7.4 channel α-subunit in hypoxic arteries, whereas Kv7.1 and Kv7.5 were unaffected. Flupirtine had no effect in normoxic lungs but reduced vascular resistance in hypoxic lungs. Moreover, oral dosing with flupirtine (30 mg/kg/day) prevented short-term in vivo hypoxia from increasing pulmonary vascular resistance and sensitizing the arteries to acute hypoxia. These findings suggest a protective role for Kv7.4 channels in the pulmonary circulation, limiting its reactivity to pressor agents and preventing hypoxia-induced pulmonary hypertension. They also provide further support for the therapeutic potential of Kv7 activators in pulmonary vascular disease.
Collapse
Affiliation(s)
- Vojtech Sedivy
- Department of Physiology, Charles University - Second Faculty of Medicine, Prague, Czech Republic; Department of Paediatrics, Charles University - Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic; and
| | - Shreena Joshi
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Youssef Ghaly
- Department of Physiology, Charles University - Second Faculty of Medicine, Prague, Czech Republic
| | - Roman Mizera
- Department of Physiology, Charles University - Second Faculty of Medicine, Prague, Czech Republic
| | - Marie Zaloudikova
- Department of Pathophysiology, Charles University - Second Faculty of Medicine, Prague, Czech Republic
| | - Sean Brennan
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Jana Novotna
- Department of Biochemistry, Charles University - Second Faculty of Medicine, Prague, Czech Republic
| | - Jan Herget
- Department of Physiology, Charles University - Second Faculty of Medicine, Prague, Czech Republic
| | - Alison M Gurney
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
54
|
Anderson UA, Carson C, Johnston L, Joshi S, Gurney AM, McCloskey KD. Functional expression of KCNQ (Kv7) channels in guinea pig bladder smooth muscle and their contribution to spontaneous activity. Br J Pharmacol 2014; 169:1290-304. [PMID: 23586426 PMCID: PMC3746117 DOI: 10.1111/bph.12210] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 03/15/2013] [Accepted: 03/26/2013] [Indexed: 12/30/2022] Open
Abstract
Background and Purpose The aim of the study was to determine whether KCNQ channels are functionally expressed in bladder smooth muscle cells (SMC) and to investigate their physiological significance in bladder contractility. Experimental Approach KCNQ channels were examined at the genetic, protein, cellular and tissue level in guinea pig bladder smooth muscle using RT-PCR, immunofluorescence, patch-clamp electrophysiology, calcium imaging, detrusor strip myography, and a panel of KCNQ activators and inhibitors. Key Results KCNQ subtypes 1–5 are expressed in bladder detrusor smooth muscle. Detrusor strips typically displayed TTX-insensitive myogenic spontaneous contractions that were increased in amplitude by the KCNQ channel inhibitors XE991, linopirdine or chromanol 293B. Contractility was inhibited by the KCNQ channel activators flupirtine or meclofenamic acid (MFA). The frequency of Ca2+-oscillations in SMC contained within bladder tissue sheets was increased by XE991. Outward currents in dispersed bladder SMC, recorded under conditions where BK and KATP currents were minimal, were significantly reduced by XE991, linopirdine, or chromanol, and enhanced by flupirtine or MFA. XE991 depolarized the cell membrane and could evoke transient depolarizations in quiescent cells. Flupirtine (20 μM) hyperpolarized the cell membrane with a simultaneous cessation of any spontaneous electrical activity. Conclusions and Implications These novel findings reveal the role of KCNQ currents in the regulation of the resting membrane potential of detrusor SMC and their important physiological function in the control of spontaneous contractility in the guinea pig bladder.
Collapse
Affiliation(s)
- U A Anderson
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | | | | | | | | | | |
Collapse
|
55
|
Fretwell LV, Woolard J. Cardiovascular responses to retigabine in conscious rats--under normotensive and hypertensive conditions. Br J Pharmacol 2014; 169:1279-89. [PMID: 23581476 DOI: 10.1111/bph.12203] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 03/08/2013] [Accepted: 03/22/2013] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Retigabine is a recently approved antiepileptic agent which activates Kv7.2-7.5 potassium channels. It is emerging that these channels have an important role in vascular regulation, but the vascular effects of retigabine in the conscious state are unknown. Hence, in the present study we assessed the regional haemodynamic responses to retigabine in conscious rats. EXPERIMENTAL APPROACH Male Sprague Dawley rats were chronically instrumented with pulsed Doppler flow probes to measure regional haemodynamic responses to retigabine under control conditions and during acute hypertension induced by infusion of angiotensin II and arginine vasopressin. Further experiments were performed, using the β-adrenoceptor antagonists CGP 20712A, ICI 118551 and propranolol, to elucidate the roles of β-adrenoceptors in the responses to retigabine in vivo and in vitro. KEY RESULTS Under normotensive conditions, retigabine induced dose-dependent hypotension and hindquarters vasodilatation, with small, transient renal and mesenteric vasodilatations. In the acutely hypertensive state, the renal and mesenteric, but not hindquarters, vasodilatations were enhanced. The response of the hindquarters vascular bed to retigabine was mediated, in part, by β₂-adrenoceptors. However, in vitro experiments confirmed that retigabine did not act as a β-adrenoceptor agonist. CONCLUSIONS AND IMPLICATIONS We demonstrated that retigabine causes regionally specific vasodilatations, which are different under normotensive and hypertensive conditions, and are, in part, mediated by β₂-adrenoceptors in some vascular beds but not in others. These results broadly support previous findings and further indicate that Kv7 channels are a potential therapeutic target for the treatment of vascular diseases associated with inappropriate vasoconstriction.
Collapse
Affiliation(s)
- L V Fretwell
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
56
|
Jepps TA, Bentzen BH, Stott JB, Povstyan OV, Sivaloganathan K, Dalby-Brown W, Greenwood IA. Vasorelaxant effects of novel Kv 7.4 channel enhancers ML213 and NS15370. Br J Pharmacol 2014; 171:4413-24. [PMID: 24909207 DOI: 10.1111/bph.12805] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/25/2014] [Accepted: 05/14/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The KCNQ-encoded voltage-gated potassium channel family (Kv 7.1-Kv 7.5) are established regulators of smooth muscle contractility, where Kv 7.4 and Kv 7.5 predominate. Various Kv 7.2-7.5 channel enhancers have been developed that have been shown to cause a vasorelaxation in both rodent and human blood vessels. Recently, two novel Kv 7 channel enhancers have been identified, ML213 and NS15370, that show increased potency, particularly on Kv 7.4 channels. The aim of this study was to characterize the effects of these novel enhancers in different rat blood vessels and compare them with Kv 7 enhancers (S-1, BMS204352, retigabine) described previously. We also sought to determine the binding sites of the new Kv 7 enhancers. KEY RESULTS Both ML213 and NS15370 relaxed segments of rat thoracic aorta, renal artery and mesenteric artery in a concentration-dependent manner. In the mesenteric artery ML213 and NS15370 displayed EC50 s that were far lower than other Kv 7 enhancers tested. Current-clamp experiments revealed that both novel enhancers, at low concentrations, caused significant hyperpolarization in mesenteric artery smooth muscle cells. In addition, we determined that the stimulatory effect of these enhancers relied on a tryptophan residue located in the S5 domain, which is the same binding site for the other Kv 7 enhancers tested in this study. CONCLUSIONS AND IMPLICATIONS This study has identified and characterized ML213 and NS15370 as potent vasorelaxants in different blood vessels, thereby highlighting these new compounds as potential therapeutics for various smooth muscle disorders.
Collapse
Affiliation(s)
- T A Jepps
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
57
|
Olschewski A, Papp R, Nagaraj C, Olschewski H. Ion channels and transporters as therapeutic targets in the pulmonary circulation. Pharmacol Ther 2014; 144:349-68. [PMID: 25108211 DOI: 10.1016/j.pharmthera.2014.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 10/24/2022]
Abstract
Pulmonary circulation is a low pressure, low resistance, high flow system. The low resting vascular tone is maintained by the concerted action of ion channels, exchangers and pumps. Under physiological as well as pathophysiological conditions, they are targets of locally secreted or circulating vasodilators and/or vasoconstrictors, leading to changes in expression or to posttranslational modifications. Both structural changes in the pulmonary arteries and a sustained increase in pulmonary vascular tone result in pulmonary vascular remodeling contributing to morbidity and mortality in pediatric and adult patients. There is increasing evidence demonstrating the pivotal role of ion channels such as K(+) and Cl(-) or transient receptor potential channels in different cell types which are thought to play a key role in vasoconstrictive remodeling. This review focuses on ion channels, exchangers and pumps in the pulmonary circulation and summarizes their putative pathophysiological as well as therapeutic role in pulmonary vascular remodeling. A better understanding of the mechanisms of their actions may allow for the development of new options for attenuating acute and chronic pulmonary vasoconstriction and remodeling treating the devastating disease pulmonary hypertension.
Collapse
Affiliation(s)
- Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Experimental Anesthesiology, Department of Anesthesia and Intensive Care Medicine, Medical University of Graz, Austria.
| | - Rita Papp
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Austria
| |
Collapse
|
58
|
Brueggemann LI, Haick JM, Cribbs LL, Byron KL. Differential activation of vascular smooth muscle Kv7.4, Kv7.5, and Kv7.4/7.5 channels by ML213 and ICA-069673. Mol Pharmacol 2014; 86:330-41. [PMID: 24944189 DOI: 10.1124/mol.114.093799] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent research suggests that smooth muscle cells express Kv7.4 and Kv7.5 voltage-activated potassium channels, which contribute to maintenance of their resting membrane voltage. New pharmacologic activators of Kv7 channels, ML213 (N-mesitybicyclo[2.2.1]heptane-2-carboxamide) and ICA-069673 N-(6-chloropyridin-3-yl)-3,4-difluorobenzamide), have been reported to discriminate among channels formed from different Kv7 subtypes. We compared the effects of ML213 and ICA-069673 on homomeric human Kv7.4, Kv7.5, and heteromeric Kv7.4/7.5 channels exogenously expressed in A7r5 vascular smooth muscle cells. We found that, despite its previous description as a selective activator of Kv7.2 and Kv7.4, ML213 significantly increased the maximum conductance of homomeric Kv7.4 and Kv7.5, as well as heteromeric Kv7.4/7.5 channels, and induced a negative shift of their activation curves. Current deactivation rates decreased in the presence of the ML213 (10 μM) for all three channel combinations. Mutants of Kv7.4 (W242L) and Kv7.5 (W235L), previously found to be insensitive to another Kv7 channel activator, retigabine, were also insensitive to ML213 (10 μM). In contrast to ML213, ICA-069673 robustly activated Kv7.4 channels but was significantly less effective on homomeric Kv7.5 channels. Heteromeric Kv7.4/7.5 channels displayed intermediate responses to ICA-069673. In each case, ICA-069673 induced a negative shift of the activation curves without significantly increasing maximal conductance. Current deactivation rates decreased in the presence of ICA-069673 in a subunit-specific manner. Kv7.4 W242L responded to ICA-069673-like wild-type Kv7.4, but a Kv7.4 F143A mutant was much less sensitive to ICA-069673. Based on these results, ML213 and ICA-069673 likely bind to different sites and are differentially selective among Kv7.4, Kv7.5, and Kv7.4/7.5 channel subtypes.
Collapse
Affiliation(s)
- Lyubov I Brueggemann
- Department of Molecular Pharmacology and Therapeutics (L.I.B., J.M.H., K.L.B.) and Cell and Molecular Physiology (L.L.C.); Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Jennifer M Haick
- Department of Molecular Pharmacology and Therapeutics (L.I.B., J.M.H., K.L.B.) and Cell and Molecular Physiology (L.L.C.); Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Leanne L Cribbs
- Department of Molecular Pharmacology and Therapeutics (L.I.B., J.M.H., K.L.B.) and Cell and Molecular Physiology (L.L.C.); Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Kenneth L Byron
- Department of Molecular Pharmacology and Therapeutics (L.I.B., J.M.H., K.L.B.) and Cell and Molecular Physiology (L.L.C.); Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| |
Collapse
|
59
|
Oliveras A, Roura-Ferrer M, Solé L, de la Cruz A, Prieto A, Etxebarria A, Manils J, Morales-Cano D, Condom E, Soler C, Cogolludo A, Valenzuela C, Villarroel A, Comes N, Felipe A. Functional assembly of Kv7.1/Kv7.5 channels with emerging properties on vascular muscle physiology. Arterioscler Thromb Vasc Biol 2014; 34:1522-30. [PMID: 24855057 DOI: 10.1161/atvbaha.114.303801] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Voltage-dependent K(+) (Kv) channels from the Kv7 family are expressed in blood vessels and contribute to cardiovascular physiology. Although Kv7 channel blockers trigger muscle contractions, Kv7 activators act as vasorelaxants. Kv7.1 and Kv7.5 are expressed in many vessels. Kv7.1 is under intense investigation because Kv7.1 blockers fail to modulate smooth muscle reactivity. In this study, we analyzed whether Kv7.1 and Kv7.5 may form functional heterotetrameric channels increasing the channel diversity in vascular smooth muscles. APPROACH AND RESULTS Kv7.1 and Kv7.5 currents elicited in arterial myocytes, oocyte, and mammalian expression systems suggest the formation of heterotetrameric complexes. Kv7.1/Kv7.5 heteromers, exhibiting different pharmacological characteristics, participate in the arterial tone. Kv7.1/Kv7.5 associations were confirmed by coimmunoprecipitation, fluorescence resonance energy transfer, and fluorescence recovery after photobleaching experiments. Kv7.1/Kv7.5 heterotetramers were highly retained at the endoplasmic reticulum. Studies in HEK-293 cells, heart, brain, and smooth and skeletal muscles demonstrated that the predominant presence of Kv7.5 stimulates release of Kv7.1/Kv7.5 oligomers out of lipid raft microdomains. Electrophysiological studies supported that KCNE1 and KCNE3 regulatory subunits further increased the channel diversity. Finally, the analysis of rat isolated myocytes and human blood vessels demonstrated that Kv7.1 and Kv7.5 exhibited a differential expression, which may lead to channel diversity. CONCLUSIONS Kv7.1 and Kv7.5 form heterotetrameric channels increasing the diversity of structures which fine-tune blood vessel reactivity. Because the lipid raft localization of ion channels is crucial for cardiovascular physiology, Kv7.1/Kv7.5 heteromers provide efficient spatial and temporal regulation of smooth muscle function. Our results shed light on the debate about the contribution of Kv7 channels to vasoconstriction and hypertension.
Collapse
Affiliation(s)
- Anna Oliveras
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Meritxell Roura-Ferrer
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Laura Solé
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Alicia de la Cruz
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Angela Prieto
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Ainhoa Etxebarria
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Joan Manils
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Daniel Morales-Cano
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Enric Condom
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Concepció Soler
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Angel Cogolludo
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Carmen Valenzuela
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Alvaro Villarroel
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Núria Comes
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.)
| | - Antonio Felipe
- From the Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain (A.O., M.R.-F., L.S., N.C., A.F.); Unidad de Biofísica, CSIC-UPV/EHU, Universidad del País Vasco, País Vasco, Spain (M.R.-F., A.E., A.V.); Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain (A.d.l.C., A.P., C.V.); Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge-Universitat de Barcelona, Barcelona, Spain (J.M., E.C., C.S.); and Departamento de Farmacología, Universidad Complutense de Madrid, Ciber Enfermedades Respiratorias (CibeRes), Madrid, Spain (A.C., D.M.-C.).
| |
Collapse
|
60
|
Schleifenbaum J, Kassmann M, Szijártó IA, Hercule HC, Tano JY, Weinert S, Heidenreich M, Pathan AR, Anistan YM, Alenina N, Rusch NJ, Bader M, Jentsch TJ, Gollasch M. Stretch-activation of angiotensin II type 1a receptors contributes to the myogenic response of mouse mesenteric and renal arteries. Circ Res 2014; 115:263-72. [PMID: 24838176 DOI: 10.1161/circresaha.115.302882] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Vascular wall stretch is the major stimulus for the myogenic response of small arteries to pressure. The molecular mechanisms are elusive, but recent findings suggest that G protein-coupled receptors can elicit a stretch response. OBJECTIVE To determine whether angiotensin II type 1 receptors (AT1R) in vascular smooth muscle cells exert mechanosensitivity and identify the downstream ion channel mediators of myogenic vasoconstriction. METHODS AND RESULTS We used mice deficient in AT1R signaling molecules and putative ion channel targets, namely AT1R, angiotensinogen, transient receptor potential channel 6 (TRPC6) channels, or several subtypes of the voltage-gated K+ (Kv7) gene family (KCNQ3, 4, or 5). We identified a mechanosensing mechanism in isolated mesenteric arteries and in the renal circulation that relies on coupling of the AT1R subtype a to a Gq/11 protein as a critical event to accomplish the myogenic response. Arterial mechanoactivation occurs after pharmacological block of AT1R and in the absence of angiotensinogen or TRPC6 channels. Activation of AT1R subtype a by osmotically induced membrane stretch suppresses an XE991-sensitive Kv channel current in patch-clamped vascular smooth muscle cells, and similar concentrations of XE991 enhance mesenteric and renal myogenic tone. Although XE991-sensitive KCNQ3, 4, and 5 channels are expressed in vascular smooth muscle cells, XE991-sensitive K+ current and myogenic contractions persist in arteries deficient in these channels. CONCLUSIONS Our results provide definitive evidence that myogenic responses of mouse mesenteric and renal arteries rely on ligand-independent, mechanoactivation of AT1R subtype a. The AT1R subtype a signal relies on an ion channel distinct from TRPC6 or KCNQ3, 4, or 5 to enact vascular smooth muscle cell activation and elevated vascular resistance.
Collapse
Affiliation(s)
- Johanna Schleifenbaum
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Mario Kassmann
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - István András Szijártó
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Hantz C Hercule
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Jean-Yves Tano
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Stefanie Weinert
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Matthias Heidenreich
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Asif R Pathan
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Yoland-Marie Anistan
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Natalia Alenina
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Nancy J Rusch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Michael Bader
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Thomas J Jentsch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Maik Gollasch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.).
| |
Collapse
|
61
|
Crosswhite P, Sun Z. Molecular mechanisms of pulmonary arterial remodeling. Mol Med 2014; 20:191-201. [PMID: 24676136 DOI: 10.2119/molmed.2013.00165] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 03/25/2014] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by a persistent elevation of pulmonary arterial pressure and pulmonary arterial remodeling with unknown etiology. Current therapeutics available for PAH are primarily directed at reducing the pulmonary blood pressure through their effects on the endothelium. It is well accepted that pulmonary arterial remodeling is primarily due to excessive pulmonary arterial smooth muscle cell (PASMC) proliferation that leads to narrowing or occlusion of the pulmonary vessels. Future effective therapeutics will be successful in reversing the vascular remodeling in the pulmonary arteries and arterioles. The purpose of this review is to provide updated information on molecular mechanisms involved in pulmonary arterial remodeling with a focus on growth factors, transcription factors, and epigenetic pathways in PASMC proliferation. In addition, this review will highlight novel therapeutic strategies for potentially reversing PASMC proliferation.
Collapse
Affiliation(s)
- Patrick Crosswhite
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
62
|
Currò D. K+ channels as potential targets for the treatment of gastrointestinal motor disorders. Eur J Pharmacol 2014; 733:97-101. [PMID: 24726846 DOI: 10.1016/j.ejphar.2014.03.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/14/2014] [Accepted: 03/27/2014] [Indexed: 12/21/2022]
Abstract
K(+) channels play important functional roles in excitable cells, as neurons and muscle cells. The activation or inhibition of K(+) channels hyperpolarizes or depolarizes the cell membrane, respectively. These effects determine in the smooth muscle decrease or increase in Ca(2+) influx through voltage-gated Ca(2+) (CaV1.2) channels and relaxation or contraction, respectively. Recent studies highlight the importance of voltage-dependent type 7 K(+) (KV7 or KCNQ) channels in regulating muscle tone and contractility in stomach and colon. KV7 channels, that include 5 subtypes (KV7.1-7.5), are activated at relatively negative potential values, close to those of the resting membrane potential for the smooth muscle cells of some segments of the gastrointestinal tract. Thus, they contribute to set the resting membrane potential and their blockade induces increase in smooth muscle contractility in stomach and colon. In addition, KV7 channel activation produces profound relaxations of gastric and colonic smooth muscle. Therefore, KV7 channel activators could be used to relax the smooth muscle and relieve symptoms in diseases such as functional dyspepsia and irritable bowel syndrome with prevalent diarrhea. The discovery of activators selective for the channel subtypes present in the smooth muscle, mainly KV7.4 and 7.5, would allow avoiding adverse cardiac and nervous system effects. A further step forward would be characterizing putative differences among the KV7 channel subtypes expressed in the various smooth muscles and synthesizing molecules specific for the gastrointestinal smooth muscle.
Collapse
Affiliation(s)
- Diego Currò
- Institute of Pharmacology, School of Medicine, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168 Rome, Italy.
| |
Collapse
|
63
|
Stott JB, Jepps TA, Greenwood IA. KV7 potassium channels: a new therapeutic target in smooth muscle disorders. Drug Discov Today 2014; 19:413-24. [DOI: 10.1016/j.drudis.2013.12.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/21/2013] [Accepted: 12/04/2013] [Indexed: 12/23/2022]
|
64
|
Brueggemann LI, Haick JM, Neuburg S, Tate S, Randhawa D, Cribbs LL, Byron KL. KCNQ (Kv7) potassium channel activators as bronchodilators: combination with a β2-adrenergic agonist enhances relaxation of rat airways. Am J Physiol Lung Cell Mol Physiol 2014; 306:L476-86. [PMID: 24441871 PMCID: PMC3949081 DOI: 10.1152/ajplung.00253.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/16/2014] [Indexed: 12/19/2022] Open
Abstract
KCNQ (Kv7 family) potassium (K(+)) channels were recently found in airway smooth muscle cells (ASMCs) from rodent and human bronchioles. In the present study, we evaluated expression of KCNQ channels and their role in constriction/relaxation of rat airways. Real-time RT-PCR analysis revealed expression of KCNQ4 > KCNQ5 > KCNQ1 > KCNQ2 > KCNQ3, and patch-clamp electrophysiology detected KCNQ currents in rat ASMCs. In precision-cut lung slices, the KCNQ channel activator retigabine induced a concentration-dependent relaxation of small bronchioles preconstricted with methacholine (MeCh; EC50 = 3.6 ± 0.3 μM). Bronchoconstriction was also attenuated in the presence of two other structurally unrelated KCNQ channel activators: zinc pyrithione (ZnPyr; 1 μM; 22 ± 7%) and 2,5-dimethylcelecoxib (10 μM; 24 ± 8%). The same three KCNQ channel activators increased KCNQ currents in ASMCs by two- to threefold. The bronchorelaxant effects of retigabine and ZnPyr were prevented by inclusion of the KCNQ channel blocker XE991. A long-acting β2-adrenergic receptor agonist, formoterol (10 nM), did not increase KCNQ current amplitude in ASMCs, but formoterol (1-1,000 nM) did induce a time- and concentration-dependent relaxation of rat airways, with a notable desensitization during a 30-min treatment or with repetitive treatments. Coadministration of retigabine (10 μM) with formoterol produced a greater peak and sustained reduction of MeCh-induced bronchoconstriction and reduced the apparent desensitization observed with formoterol alone. Our findings support a role for KCNQ K(+) channels in the regulation of airway diameter. A combination of a β2-adrenergic receptor agonist with a KCNQ channel activator may improve bronchodilator therapy.
Collapse
Affiliation(s)
- Lioubov I Brueggemann
- Dept. of Molecular Pharmacology & Therapeutics, Loyola Univ. Chicago, Stritch School of Medicine, 2160 S. First Ave., Bldg. 102, Rm. 3634, Maywood, IL 60153.
| | | | | | | | | | | | | |
Collapse
|
65
|
Jepps TA, Olesen SP, Greenwood IA. One man's side effect is another man's therapeutic opportunity: targeting Kv7 channels in smooth muscle disorders. Br J Pharmacol 2014; 168:19-27. [PMID: 22880633 DOI: 10.1111/j.1476-5381.2012.02133.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Retigabine is a first in class anticonvulsant that has recently undergone clinical trials to test its efficacy in epileptic patients. Retigabine's novel mechanism of action - activating Kv7 channels - suppresses neuronal activity to prevent seizure generation by hyperpolarizing the membrane potential and suppressing depolarizing surges. However, Kv7 channels are not expressed exclusively in neurones and data generated over the last decade have shown that Kv7 channels play a key role in various smooth muscle systems of the body. This review discusses the potential of targeting Kv7 channels in the smooth muscle to treat diseases such as hypertension, bladder instability, constipation and preterm labour.
Collapse
Affiliation(s)
- T A Jepps
- Division of Biomedical Sciences, St George's, University of London, Cranmer Terrace, UK
| | | | | |
Collapse
|
66
|
Makino A, Firth AL, Yuan JXJ. Endothelial and smooth muscle cell ion channels in pulmonary vasoconstriction and vascular remodeling. Compr Physiol 2013; 1:1555-602. [PMID: 23733654 DOI: 10.1002/cphy.c100023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pulmonary circulation is a low resistance and low pressure system. Sustained pulmonary vasoconstriction and excessive vascular remodeling often occur under pathophysiological conditions such as in patients with pulmonary hypertension. Pulmonary vasoconstriction is a consequence of smooth muscle contraction. Many factors released from the endothelium contribute to regulating pulmonary vascular tone, while the extracellular matrix in the adventitia is the major determinant of vascular wall compliance. Pulmonary vascular remodeling is characterized by adventitial and medial hypertrophy due to fibroblast and smooth muscle cell proliferation, neointimal proliferation, intimal, and plexiform lesions that obliterate the lumen, muscularization of precapillary arterioles, and in situ thrombosis. A rise in cytosolic free Ca(2+) concentration ([Ca(2+)]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction, while increased release of mitogenic factors, upregulation (or downregulation) of ion channels and transporters, and abnormalities in intracellular signaling cascades are key to the remodeling of the pulmonary vasculature. Changes in the expression, function, and regulation of ion channels in PASMC and pulmonary arterial endothelial cells play an important role in the regulation of vascular tone and development of vascular remodeling. This article will focus on describing the ion channels and transporters that are involved in the regulation of pulmonary vascular function and structure and illustrating the potential pathogenic role of ion channels and transporters in the development of pulmonary vascular disease.
Collapse
Affiliation(s)
- Ayako Makino
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
67
|
Brueggemann LI, Mackie AR, Cribbs LL, Freda J, Tripathi A, Majetschak M, Byron KL. Differential protein kinase C-dependent modulation of Kv7.4 and Kv7.5 subunits of vascular Kv7 channels. J Biol Chem 2013; 289:2099-111. [PMID: 24297175 DOI: 10.1074/jbc.m113.527820] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The Kv7 family (Kv7.1-7.5) of voltage-activated potassium channels contributes to the maintenance of resting membrane potential in excitable cells. Previously, we provided pharmacological and electrophysiological evidence that Kv7.4 and Kv7.5 form predominantly heteromeric channels and that Kv7 activity is regulated by protein kinase C (PKC) in response to vasoconstrictors in vascular smooth muscle cells. Direct evidence for Kv7.4/7.5 heteromer formation, however, is lacking. Furthermore, it remains to be determined whether both subunits are regulated by PKC. Utilizing proximity ligation assays to visualize single molecule interactions, we now show that Kv7.4/Kv.7.5 heteromers are endogenously expressed in vascular smooth muscle cells. Introduction of dominant-negative Kv7.4 and Kv7.5 subunits in mesenteric artery myocytes reduced endogenous Kv7 currents by 84 and 76%, respectively. Expression of an inducible protein kinase Cα (PKCα) translocation system revealed that PKCα activation is sufficient to suppress endogenous Kv7 currents in A7r5 rat aortic and mesenteric artery smooth muscle cells. Arginine vasopressin (100 and 500 pm) and the PKC activator phorbol 12-myristate 13-acetate (1 nm) each inhibited human (h) Kv7.5 and hKv7.4/7.5, but not hKv7.4 channels expressed in A7r5 cells. A decrease in hKv7.5 and hKv7.4/7.5 current densities was associated with an increase in PKC-dependent phosphorylation of the channel proteins. These findings provide further evidence for a differential regulation of Kv7.4 and Kv7.5 channel subunits by PKC-dependent phosphorylation and new mechanistic insights into the role of heteromeric subunit assembly for regulation of vascular Kv7 channels.
Collapse
|
68
|
Evseev AI, Semenov I, Archer CR, Medina JL, Dube PH, Shapiro MS, Brenner R. Functional effects of KCNQ K(+) channels in airway smooth muscle. Front Physiol 2013; 4:277. [PMID: 24109455 PMCID: PMC3791379 DOI: 10.3389/fphys.2013.00277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/13/2013] [Indexed: 12/30/2022] Open
Abstract
KCNQ (Kv7) channels underlie a voltage-gated K+ current best known for control of neuronal excitability, and its inhibition by Gq/11-coupled, muscarinic signaling. Studies have indicated expression of KCNQ channels in airway smooth muscle (ASM), a tissue that is predominantly regulated by muscarinic receptor signaling. Therefore, we investigated the function of KCNQ channels in rodent ASM and their interplay with Gq/11-coupled M3 muscarinic receptors. Perforated-patch clamp of dissociated ASM cells detected a K+ current inhibited by the KCNQ antagonist, XE991, and augmented by the specific agonist, flupirtine. KCNQ channels begin to activate at voltages near resting potentials for ASM cells, and indeed XE991 depolarized resting membrane potentials. Muscarinic receptor activation inhibited KCNQ current weakly (~20%) at concentrations half-maximal for contractions. Thus, we were surprised to see that KCNQ had no affect on membrane voltage or muscle contractility following muscarinic activation. Further, M3 receptor-specific antagonist J104129 fumarate alone did not reveal KCNQ effects on muscarinic evoked depolarization or contractility. However, a role for KCNQ channels was revealed when BK-K+ channel activities are reduced. While KCNQ channels do control resting potentials, they appear to play a redundant role with BK calcium-activated K+ channels during ASM muscarinic signaling. In contrast to effect of antagonist, we observe that KCNQ agonist flupirtine caused a significant hyperpolarization and reduced contraction in vitro irrespective of muscarinic activation. Using non-invasive whole animal plethysmography, the clinically approved KCNQ agonist retigabine caused a transient reduction in indexes of airway resistance in both wild type and BK β1 knockout (KO) mice treated with the muscarinic agonist. These findings indicate that KCNQ channels can be recruited via agonists to oppose muscarinic evoked contractions and may be of therapeutic value as bronchodilators.
Collapse
Affiliation(s)
- Alexey I Evseev
- Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Affiliation(s)
- Maik Gollasch
- Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
70
|
Vascular KCNQ (Kv7) potassium channels as common signaling intermediates and therapeutic targets in cerebral vasospasm. J Cardiovasc Pharmacol 2013; 61:51-62. [PMID: 23107868 DOI: 10.1097/fjc.0b013e3182771708] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cerebral vasospasm after subarachnoid hemorrhage (SAH) is characterized by prolonged severe constriction of the basilar artery, which often leads to ischemic brain damage. Locally elevated concentrations of spasmogenic substances induce persistent depolarization of myocytes in the basilar artery, leading to continuous influx of calcium (Ca) through voltage-sensitive Ca channels and myocyte contraction. Potassium (K) channel openers may have therapeutic utility to oppose membrane depolarization, dilate the arteries, and reduce ischemia. Here, we examined the involvement of vascular Kv7 K channels in the pathogenesis of cerebral vasospasm and tested whether Kv7 channel openers are effective therapeutic agents in a rat model of SAH. Patch-clamp experiments revealed that 3 different spasmogens (serotonin, endothelin, and vasopressin) suppressed Kv7 currents and depolarized freshly isolated rat basilar artery myocytes. These effects were significantly reduced in the presence of a Kv7 channel opener, retigabine. Retigabine (10 μM) also significantly blocked L-type Ca channels, reducing peak inward currents by >50%. In the presence of a selective Kv7 channel blocker, XE991, the spasmogens did not produce additive constriction responses measured using pressure myography. Kv7 channel openers (retigabine or celecoxib) significantly attenuated basilar artery spasm in rats with experimentally induced SAH. In conclusion, we identify Kv7 channels as common targets of vasoconstrictor spasmogens and as candidates for therapeutic intervention for cerebral vasospasm.
Collapse
|
71
|
Joseph BK, Thakali KM, Moore CL, Rhee SW. Ion channel remodeling in vascular smooth muscle during hypertension: Implications for novel therapeutic approaches. Pharmacol Res 2013; 70:126-38. [PMID: 23376354 PMCID: PMC3607210 DOI: 10.1016/j.phrs.2013.01.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 01/10/2013] [Accepted: 01/17/2013] [Indexed: 02/07/2023]
Abstract
Ion channels are multimeric, transmembrane proteins that selectively mediate ion flux across the plasma membrane in a variety of cells including vascular smooth muscle cells (VSMCs). The dynamic interplay of Ca(2+) and K(+) channels on the plasma membrane of VSMCs plays a pivotal role in modulating the vascular tone of small arteries and arterioles. The abnormally-elevated arterial tone observed in hypertension thus points to an aberrant expression and function of Ca(2+) and K(+) channels in the VSMCs. In this short review, we focus on the three well-studied ion channels in VSMCs, namely the L-type Ca(2+) (CaV1.2) channels, the voltage-gated K(+) (KV) channels, and the large-conductance Ca(2+)-activated K(+) (BK) channels. First, we provide a brief overview on the physiological role of vascular CaV1.2, KV and BK channels in regulating arterial tone. Second, we discuss the current understanding of the expression changes and regulation of CaV1.2, KV and BK channels in the vasculature during hypertension. Third, based on available proof-of-concept studies, we describe the potential therapeutic approaches targeting these vascular ion channels in order to restore blood pressure to normotensive levels.
Collapse
Affiliation(s)
- Biny K Joseph
- Venenum Biodesign, 8 Black Forest Road, Hamilton, NJ 08691, USA
| | | | | | | |
Collapse
|
72
|
Hosseinzadeh Z, Sopjani M, Pakladok T, Bhavsar SK, Lang F. Downregulation of KCNQ4 by Janus kinase 2. J Membr Biol 2013; 246:335-41. [PMID: 23543186 DOI: 10.1007/s00232-013-9537-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 03/16/2013] [Indexed: 12/19/2022]
Abstract
Janus kinase-2 (JAK2) participates in the signaling of several hormones, growth factors and cytokines. Further stimulators of JAK2 include osmotic cell shrinkage, and the kinase activates the cell volume regulatory Na(+)/H(+) exchanger. The kinase may thus participate in cell volume regulation. Cell shrinkage is known to inhibit K(+) channels. Volume-regulatory K(+) channels include the voltage-gated K(+) channel KCNQ4. The present study explored the effect of JAK2 on KCNQ4 channel activity. KCNQ4 was expressed in Xenopus oocytes with or without wild-type JAK2, constitutively active (V617F)JAK2 or inactive (K882E)JAK2; and cell membrane conductance was determined by dual-electrode voltage clamp. Expression of KCNQ4 was followed by the appearance of voltage-gated K(+) conductance. Coexpression of JAK2 or of (V617F)JAK2, but not of (K882E)JAK2, resulted in a significant decrease in conductance. Treatment of KCNQ4 and JAK2 coexpressing oocytes with the JAK2 inhibitor AG490 (40 μM) was followed by an increase in conductance. Treatment of KCNQ4 expressing oocytes with brefeldin A (5 μM) was followed by a decrease in conductance, which was similar in oocytes expressing KCNQ4 together with JAK2 as in oocytes expressing KCNQ4 alone. Thus, JAK2 apparently does not accelerate channel protein retrieval from the cell membrane. In conclusion, JAK2 downregulates KCNQ4 activity and thus counteracts K(+) exit, an effect which may contribute to cell volume regulation.
Collapse
|
73
|
Flupirtine, a re-discovered drug, revisited. Inflamm Res 2013; 62:251-8. [PMID: 23322112 DOI: 10.1007/s00011-013-0592-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/20/2012] [Accepted: 01/02/2013] [Indexed: 12/15/2022] Open
Abstract
Flupirtine was developed long before K(V)7 (KCNQ) channels were known. However, it was clear from the beginning that flupirtine is neither an opioid nor a nonsteroidal anti-inflammatory analgesic. Its unique muscle relaxing activity was discovered by serendipity. In the meantime, broad and intensive research has resulted in a partial clarification of its mode of action. Flupirtine is the first therapeutically used K(V)7 channel activator with additional GABA(A)ergic mechanisms and thus the first representative of a novel class of analgesics. The presently accepted main mode of its action, potassium K(V)7 (KCNQ) channel activation, opens a series of further therapeutic possibilities. One of them has now been realized: its back-up compound, the bioisostere retigabine, has been approved for the treatment of epilepsy.
Collapse
|
74
|
Chadha PS, Zunke F, Davis AJ, Jepps TA, Linders JTM, Schwake M, Towart R, Greenwood IA. Pharmacological dissection of K(v)7.1 channels in systemic and pulmonary arteries. Br J Pharmacol 2012; 166:1377-87. [PMID: 22251082 DOI: 10.1111/j.1476-5381.2012.01863.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to characterize the functional impact of KCNQ1-encoded voltage-dependent potassium channels (K(v)7.1) in the vasculature. EXPERIMENTAL APPROACH Mesenteric arteries, intrapulmonary arteries and thoracic aortae were isolated from adult rats. K(v)7.1 channel expression was established by fluorescence immunocytochemistry. Wire myography determined functionality of these channels in response to selective blockers and activators. Xenopus oocytes expressing K(v)7.1 channels were used to assess the effectiveness of selective K(v)7.1 channel blockers. KEY RESULTS K(v)7.1 channels were identified in arterial myocytes by immunocytochemistry. K(v)7.1 blockers HMR1556, L-768,673 (10 µM) and JNJ39490282 (JNJ282; 1 µM) had no contractile effects in arteries, whereas the pan-K(v)7 channel blocker linopirdine (10 µM) evoked robust contractions. Application of two compounds purported to activate K(v)7.1 channels, L-364 373 (R-L3) and mefenamic acid, relaxed mesenteric arteries preconstricted by methoxamine. These responses were reversed by HMR1556 or L-768,673 but not JNJ282. Similar effects were observed in the thoracic aorta and intrapulmonary arteries. CONCLUSIONS AND IMPLICATIONS In contrast to previous assumptions, K(v)7.1 channels expressed in arterial myocytes are functional ion channels. Although these channels do not appear to contribute to resting vascular tone, K(v)7.1 activators were effective vasorelaxants.
Collapse
Affiliation(s)
- Preet S Chadha
- Division of Biomedical Sciences: Ion Channels and Cell Signalling Group, St George's, University of London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Wrobel E, Tapken D, Seebohm G. The KCNE Tango - How KCNE1 Interacts with Kv7.1. Front Pharmacol 2012; 3:142. [PMID: 22876232 PMCID: PMC3410610 DOI: 10.3389/fphar.2012.00142] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/29/2012] [Indexed: 12/23/2022] Open
Abstract
The classical tango is a dance characterized by a 2/4 or 4/4 rhythm in which the partners dance in a coordinated way, allowing dynamic contact. There is a surprising similarity between the tango and how KCNE β-subunits "dance" to the fast rhythm of the cell with their partners from the Kv channel family. The five KCNE β-subunits interact with several members of the Kv channels, thereby modifying channel gating via the interaction of their single transmembrane-spanning segment, the extracellular amino terminus, and/or the intracellular carboxy terminus with the Kv α-subunit. Best studied is the molecular basis of interactions between KCNE1 and Kv7.1, which, together, supposedly form the native cardiac I(Ks) channel. Here we review the current knowledge about functional and molecular interactions of KCNE1 with Kv7.1 and try to summarize and interpret the tango of the KCNEs.
Collapse
Affiliation(s)
- Eva Wrobel
- Cation Channel Group, Department of Biochemistry I, Faculty of Chemistry and Biochemistry, Ruhr University Bochum Bochum, Germany
| | | | | |
Collapse
|
76
|
Ciliberto MA, Weisenberg JL, Wong M. Clinical utility, safety, and tolerability of ezogabine (retigabine) in the treatment of epilepsy. DRUG HEALTHCARE AND PATIENT SAFETY 2012; 4:81-6. [PMID: 22888276 PMCID: PMC3413039 DOI: 10.2147/dhps.s28814] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
One-third of patients with epilepsy continue to have seizures despite current treatments, indicating the need for better antiseizure medications with novel mechanisms of action. Ezogabine (retigabine) has recently been approved for adjunctive treatment of partial-onset seizures in adult patients with epilepsy. Ezogabine utilizes a novel mechanism of action, involving activation of specific potassium channels. The most common side effects of ezogabine are shared by most antiseizure medications and primarily consist of central nervous system (CNS) symptoms, such as somnolence, dizziness, confusion, and fatigue. In addition, a small percentage of patients on ezogabine experience a unique adverse effect affecting the bladder, which results in urinary hesitancy; thus, patients on ezogabine should be monitored carefully for potential urological symptoms. Overall, ezogabine appears to be well tolerated and represents a reasonable new option for treating patients with intractable epilepsy.
Collapse
Affiliation(s)
- Michael A Ciliberto
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | | | | |
Collapse
|
77
|
Chadha PS, Zunke F, Zhu HL, Davis AJ, Jepps TA, Olesen SP, Cole WC, Moffatt JD, Greenwood IA. Reduced KCNQ4-encoded voltage-dependent potassium channel activity underlies impaired β-adrenoceptor-mediated relaxation of renal arteries in hypertension. Hypertension 2012; 59:877-84. [PMID: 22353613 DOI: 10.1161/hypertensionaha.111.187427] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
KCNQ4-encoded voltage-dependent potassium (Kv7.4) channels are important regulators of vascular tone that are severely compromised in models of hypertension. However, there is no information as to the role of these channels in responses to endogenous vasodilators. We used a molecular knockdown strategy, as well as pharmacological tools, to examine the hypothesis that Kv7.4 channels contribute to β-adrenoceptor-mediated vasodilation in the renal vasculature and underlie the vascular deficit in spontaneously hypertensive rats. Quantitative PCR and immunohistochemistry confirmed gene and protein expression of KCNQ1, KCNQ3, KCNQ4, KCNQ5, and Kv7.1, Kv7.4, and Kv7.5 in rat renal artery. Isoproterenol produced concentration-dependent relaxation of precontracted renal arteries and increased Kv7 channel currents in isolated smooth muscle cells. Application of the Kv7 blocker linopirdine attenuated isoproterenol-induced relaxation and current. Isoproterenol-induced relaxations were also reduced in arteries incubated with small interference RNAs targeted to KCNQ4 that produced a ≈60% decrease in Kv7.4 protein level. Relaxation to isoproterenol and the Kv7 activator S-1 were abolished in arteries from spontaneously hypertensive rats, which was associated with ≈60% decrease in Kv7.4 abundance. This study provides the first evidence that Kv7 channels contribute to β-adrenoceptor-mediated vasodilation in the renal vasculature and that abrogation of Kv7.4 channels is strongly implicated in the impaired β-adrenoceptor pathway in spontaneously hypertensive rats. These findings may provide a novel pathogenic link between arterial dysfunction and hypertension.
Collapse
Affiliation(s)
- Preet S Chadha
- Division of Biomedical Sciences, Pharmacology and Cell Physiology Research Group, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Kuhr FK, Smith KA, Song MY, Levitan I, Yuan JXJ. New mechanisms of pulmonary arterial hypertension: role of Ca²⁺ signaling. Am J Physiol Heart Circ Physiol 2012; 302:H1546-62. [PMID: 22245772 DOI: 10.1152/ajpheart.00944.2011] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease that usually culminates in right heart failure and death if left untreated. Although there have been substantial improvements in our understanding and significant advances in the management of this disease, there is a grim prognosis for patients in the advanced stages of PAH. A major cause of PAH is increased pulmonary vascular resistance, which results from sustained vasoconstriction, excessive pulmonary vascular remodeling, in situ thrombosis, and increased pulmonary vascular stiffness. In addition to other signal transduction pathways, Ca(2+) signaling in pulmonary artery smooth muscle cells (PASMCs) plays a central role in the development and progression of PAH because of its involvement in both vasoconstriction, through its pivotal effect of PASMC contraction, and vascular remodeling, through its stimulatory effect on PASMC proliferation. Altered expression, function, and regulation of ion channels and transporters in PASMCs contribute to an increased cytosolic Ca(2+) concentration and enhanced Ca(2+) signaling in patients with PAH. This review will focus on the potential pathogenic role of Ca(2+) mobilization, regulation, and signaling in the development and progression of PAH.
Collapse
Affiliation(s)
- Frank K Kuhr
- Section of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
79
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
80
|
Yoo HY, Park SJ, Seo EY, Park KS, Han JA, Kim KS, Shin DH, Earm YE, Zhang YH, Kim SJ. Role of thromboxane A2-activated nonselective cation channels in hypoxic pulmonary vasoconstriction of rat. Am J Physiol Cell Physiol 2012; 302:C307-17. [DOI: 10.1152/ajpcell.00153.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypoxia-induced pulmonary vasoconstriction (HPV) is critical for matching of ventilation/perfusion in lungs. Although hypoxic inhibition of K+ channels has been a leading hypothesis for depolarization of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia, pharmacological inhibition of K+ channels does not induce significant contraction in rat pulmonary arteries. Because a partial contraction by thromboxane A2 (TXA2) is required for induction of HPV, we hypothesize that TXA2 receptor (TP) stimulation might activate depolarizing nonselective cation channels (NSCs). Consistently, we found that 5–10 nM U46619, a stable agonist for TP, was indispensible for contraction of rat pulmonary arteries by 4-aminopyridine, a blocker of voltage-gated K+ channel (Kv). Whole cell voltage clamp with rat PASMC revealed that U46619 induced a NSC current ( INSC,TXA2) with weakly outward rectifying current-voltage relation. INSC,TXA2 was blocked by ruthenium red (RR), an antagonist of the transient receptor potential vanilloid-related channel (TRPV) subfamily. 2-Aminoethoxydiphenyl borate, an agonist for TRPV1–3, consistently activated NSC channels in PASMCs. In contrast, agonists for TRPV1 (capsaicin), TRPV3 (camphor), or TRPV4 (α-PDD) rarely induced an increase in the membrane conductance of PASMCs. RT-PCR analysis showed the expression of transcripts for TRPV2 and -4 in rat PASMCs. Finally, it was confirmed that pretreatment with RR largely inhibited HPV in the presence of U46619. The pretreatment with agonists for TRPV1 (capsaicin) and TRPV4 (α-PDD) was ineffective as pretone agents for HPV. Taken together, it is suggested that the concerted effects of INSC,TXA2 activation and Kv inhibition under hypoxia induce membrane depolarization sufficient for HPV. TRPV2 is carefully suggested as the TXA2-activated NSC in rat PASMC.
Collapse
Affiliation(s)
- Hae Young Yoo
- Department of Physiology, Seoul National University College of Medicine
| | - Su Jung Park
- Department of Physiology, Seoul National University College of Medicine
| | - Eun-Young Seo
- Department of Physiology, Seoul National University College of Medicine
| | - Kyung Sun Park
- Division of Intergrative Biosciences and Biotechnology, POSTECH; and
| | - Jung-A. Han
- Department of Physiology, Seoul National University College of Medicine
| | - Kyung Soo Kim
- Department of Physiology, Seoul National University College of Medicine
| | - Dong Hoon Shin
- Department of Physiology, Seoul National University College of Medicine
| | - Yung E. Earm
- Department of Physiology, Seoul National University College of Medicine
| | - Yin-Hua Zhang
- Department of Physiology, Seoul National University College of Medicine
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine
- Ischemic/Hypoxic Disease Institute, Kidney Research Institute, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
81
|
Brueggemann LI, Kakad PP, Love RB, Solway J, Dowell ML, Cribbs LL, Byron KL. Kv7 potassium channels in airway smooth muscle cells: signal transduction intermediates and pharmacological targets for bronchodilator therapy. Am J Physiol Lung Cell Mol Physiol 2011; 302:L120-32. [PMID: 21964407 DOI: 10.1152/ajplung.00194.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Expression and function of Kv7 (KCNQ) voltage-activated potassium channels in guinea pig and human airway smooth muscle cells (ASMCs) were investigated by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), patch-clamp electrophysiology, and precision-cut lung slices. qRT-PCR revealed expression of multiple KCNQ genes in both guinea pig and human ASMCs. Currents with electrophysiological and pharmacological characteristics of Kv7 currents were measured in freshly isolated guinea pig and human ASMCs. In guinea pig ASMCs, Kv7 currents were significantly suppressed by application of the bronchoconstrictor agonists methacholine (100 nM) or histamine (30 μM), but current amplitudes were restored by addition of a Kv7 channel activator, flupirtine (10 μM). Kv7 currents in guinea pig ASMCs were also significantly enhanced by another Kv7.2-7.5 channel activator, retigabine, and by celecoxib and 2,5-dimethyl celecoxib. In precision-cut human lung slices, constriction of airways by histamine was significantly reduced in the presence of flupirtine. Kv7 currents in both guinea pig and human ASMCs were inhibited by the Kv7 channel blocker XE991. In human lung slices, XE991 induced robust airway constriction, which was completely reversed by addition of the calcium channel blocker verapamil. These findings suggest that Kv7 channels in ASMCs play an essential role in the regulation of airway diameter and may be targeted pharmacologically to relieve airway hyperconstriction induced by elevated concentrations of bronchoconstrictor agonists.
Collapse
Affiliation(s)
- Lioubov I Brueggemann
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA
| | | | | | | | | | | | | |
Collapse
|
82
|
Svalø J, Hansen HH, Rønn LCB, Sheykhzade M, Munro G, Rode F. Kv7 Positive Modulators Reduce Detrusor Overactivity and Increase Bladder Capacity in Rats. Basic Clin Pharmacol Toxicol 2011; 110:145-53. [DOI: 10.1111/j.1742-7843.2011.00765.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
83
|
Chadha PS, Greenwood IA, Zhong XZ, Cole WC. Letter to the Editor re Mani et al. Br J Pharmacol 2011; 164:250-1; author reply 252-3. [DOI: 10.1111/j.1476-5381.2011.01454.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
84
|
Mani BK, Brueggemann LI, Cribbs LL, Byron KL. Activation of vascular KCNQ (Kv7) potassium channels reverses spasmogen-induced constrictor responses in rat basilar artery. Br J Pharmacol 2011; 164:237-49. [PMID: 21323904 PMCID: PMC3174403 DOI: 10.1111/j.1476-5381.2011.01273.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/03/2010] [Accepted: 01/12/2011] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Cerebral vasospasm is the persistent constriction of large conduit arteries in the base of the brain. This pathologically sustained contraction of the arterial myocytes has been attributed to locally elevated concentrations of vasoconstrictor agonists (spasmogens). We assessed the presence and function of KCNQ (K(v) 7) potassium channels in rat basilar artery myocytes, and determined the efficacy of K(v) 7 channel activators in relieving spasmogen-induced basilar artery constriction. EXPERIMENTAL APPROACH Expression and function of K(v) 7 channels in freshly isolated basilar artery myocytes were evaluated by reverse transcriptase polymerase chain reaction and whole-cell electrophysiological techniques. Functional responses to K(v) 7 channel modulators were studied in intact artery segments using pressure myography. KEY RESULTS All five mammalian KCNQ subtypes (KCNQ1-5) were detected in the myocytes. K(v) currents were attributed to K(v) 7 channel activity based on their voltage dependence of activation (V(0.5) ∼-34 mV), lack of inactivation, enhancement by flupirtine (a selective K(v) 7 channel activator) and inhibition by 10,10-bis(pyridin-4-ylmethyl)anthracen-9-one (XE991; a selective K(v) 7 channel blocker). XE991 depolarized the myocytes and constricted intact basilar arteries. Celecoxib, a clinically used anti-inflammatory drug, not only enhanced K(v) 7 currents but also inhibited voltage-sensitive Ca(2+) currents. In arteries pre-constricted with spasmogens, both celecoxib and flupirtine were more effective in dilating artery segments than was nimodipine, a selective L-type Ca(2+) channel blocker. CONCLUSIONS AND IMPLICATIONS K(v) 7 channels are important determinants of basilar artery contractile status. Targeting the K(v) 7 channels using flupirtine or celecoxib could provide a novel strategy to relieve basilar artery constriction in patients with cerebral vasospasm. LINKED ARTICLES To view two letters to the Editor regarding this article visit http://dx.doi.org/10.1111/j.1476-5381.2011.01454.x and http://dx.doi.org/10.1111/j.1476-5381.2011.01457.x.
Collapse
Affiliation(s)
- Bharath K Mani
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | | | | | | |
Collapse
|
85
|
Jepps TA, Chadha PS, Davis AJ, Harhun MI, Cockerill GW, Olesen SP, Hansen RS, Greenwood IA. Downregulation of Kv7.4 channel activity in primary and secondary hypertension. Circulation 2011; 124:602-11. [PMID: 21747056 DOI: 10.1161/circulationaha.111.032136] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Voltage-gated potassium (K(+)) channels encoded by KCNQ genes (Kv7 channels) have been identified in various rodent and human blood vessels as key regulators of vascular tone; however, nothing is known about the functional impact of these channels in vascular disease. We ascertained the effect of 3 structurally different activators of Kv7.2 through Kv7.5 channels (BMS-204352, S-1, and retigabine) on blood vessels from normotensive and hypertensive animals. METHODS AND RESULTS Precontracted thoracic aorta and mesenteric artery segments from normotensive rats were relaxed by all 3 Kv7 activators, with potencies of BMS-204352=S-1>retigabine. We also tested these agents in the coronary circulation using the Langendorff heart preparation. BMS-204352 and S-1 dose dependently increased coronary perfusion at concentrations between 0.1 and 10 μmol/L, whereas retigabine was effective at 1 to 10 μmol/L. In addition, S-1 increased K(+) currents in isolated mesenteric artery myocytes. The ability of these agents to relax precontracted vessels, increase coronary flow, or augment K(+) currents was impaired considerably in tissues isolated from spontaneously hypertensive rats (SHRs). Of the 5 KCNQ genes, only the expression of KCNQ4 was reduced (≈3.7 fold) in SHRs aorta. Kv7.4 protein levels were ≈50% lower in aortas and mesenteric arteries from spontaneously hypertensive rats compared with normotensive vessels. A similar attenuated response to S-1 and decreased Kv7.4 were observed in mesenteric arteries from mice made hypertensive by angiotensin II infusion compared with normotensive controls. CONCLUSIONS In 2 different rat and mouse models of hypertension, the functional impact of Kv7 channels was dramatically downregulated.
Collapse
Affiliation(s)
- Thomas A Jepps
- Division of Biomedical Sciences, St. George's, University of London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Ipavec V, Martire M, Barrese V, Taglialatela M, Currò D. KV7 channels regulate muscle tone and nonadrenergic noncholinergic relaxation of the rat gastric fundus. Pharmacol Res 2011; 64:397-409. [PMID: 21740972 PMCID: PMC3178758 DOI: 10.1016/j.phrs.2011.06.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/14/2011] [Accepted: 06/21/2011] [Indexed: 12/30/2022]
Abstract
Voltage-dependent type 7 K+ (KV7) channels play important physiological roles in neurons and muscle cells. The aims of the present study were to investigate the motor effects of KV7 channel modulators in the rat gastric fundus and the expression of KV7 channels in this tissue. Muscle tone and electrical field stimulation (EFS)-evoked relaxations of precontracted longitudinal muscle strips of the rat gastric fundus were investigated under nonadrenergic noncholinergic conditions by organ bath studies. Gene expression was studied by real-time PCR and tissue localization of channels was investigated by immunohistochemistry. The KV7 channel blocker XE-991 induced concentration-dependent contractions, with mean pD2 and Emax of 5.4 and 48% of the maximal U46619-induced contraction, respectively. The KV7 channel activators retigabine and flupirtine concentration-dependently relaxed U46619-precontracted strips, with pD2s of 4.7 and 4.4 and Emax of 93% and 91% of the maximal relaxation induced by papaverine, respectively. XE-991 concentration-dependently inhibited retigabine-induced relaxation with a pIC50 of 6.2. XE-991 and DMP-543, another KV7 channel blocker, increased by 13-25% or reduced by 11-21% the relaxations evoked by low- or high-frequency EFS, respectively. XE-991 also reduced the relaxation induced by vasoactive intestinal polypeptide (VIP) by 33% of controls. Transcripts encoded by all KV7 genes were detected in the fundus, with 7.4 and 7.5 showing the highest expression levels. KV7.4 and 7.5 channels were visualized by confocal immunofluorescence in both circular and longitudinal muscle layers. In conclusion, in the rat proximal stomach, KV7 channels appear to contribute to the resting muscle tone and to VIP- and high-frequency EFS-induced relaxation. KV7 channel activators could be useful relaxant agents of the gastric smooth muscle.
Collapse
Affiliation(s)
- V Ipavec
- Institute of Pharmacology, School of Medicine, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168 Rome, Italy
| | | | | | | | | |
Collapse
|
87
|
Manoury B, Lamalle C, Oliveira R, Reid J, Gurney AM. Contractile and electrophysiological properties of pulmonary artery smooth muscle are not altered in TASK-1 knockout mice. J Physiol 2011; 589:3231-46. [PMID: 21486782 DOI: 10.1113/jphysiol.2011.206748] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The acid-sensitive, two-pore domain K+ channel, TASK-1, contributes to the background K+ conductance and membrane potential (Em) of rat and human pulmonary artery smooth muscle cells (PASMCs), but its role in regulating tone remains elusive. This study aimed to clarify the role of TASK-1 by determining the functional properties of pulmonary artery (PA) from mice in which the TASK-1 gene was deleted (TASK-1/3 KO), in comparison with wild-type (WT) C57BL/6 controls. Small vessel wire myography was used to measure isometric tension developed by intact PA. Em and currents were recorded from freshly isolated PASMCs using the perforated patch-clamp technique. Reverse transcription-polymerase chain reaction (RT-PCR) was used to estimate K+ channel expression. We could find no difference between PA from WT and TASK-1/3 KO mice. They showed similar constrictor responses to a range of agonists and K+ concentrations, the K+ channel blockers 4-aminopyridine, tetraethylammonium ions and XE991. Treprostinil, proposed to dilate by activating TASK-1, was just as effective in TASK-1/3 KO arteries. Blocking Ca2+ influx with nifedipine (1 μM) or levcromakalim (10 μM) had no effect on resting tone in either strain. The resting Em of PASMCs and its responses to K+ channel blockers were unchanged in TASK-1/3 KO mice as were voltage-activated K+ currents, including the non-inactivating K+ current (IKN) measured at 0 mV. The Em was, however, depolarised in comparison with other species.Mouse IKN was much smaller than in rat and showed no sensitivity to pH. The results imply that TASK-1 does not form a functional channel in mouse PASMCs.
Collapse
Affiliation(s)
- Boris Manoury
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
88
|
Mani BK, Byron KL. Vascular KCNQ channels in humans: the sub-threshold brake that regulates vascular tone? Br J Pharmacol 2011; 162:38-41. [PMID: 21188789 DOI: 10.1111/j.1476-5381.2010.01065.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Contraction of arterial smooth muscle cells results in vasoconstriction, which in turn reduces blood flow and increases blood pressure. There has been a great deal of interest in understanding the ionic mechanisms that regulate smooth muscle contraction, in part because ion channels represent potential pharmacological targets for therapies directed towards cardiovascular diseases and other conditions. Potassium channels have been recognized for their roles in maintaining or stabilizing negative membrane voltages. Activation of potassium channels opposes opening of voltage-sensitive calcium channels which conduct calcium ions into the smooth muscle cells to stimulate contraction. KCNQ potassium channels were recently discovered in arterial smooth muscle cells from rats and mice. These channels have distinctive pharmacological and biophysical characteristics that have led them to be implicated as important regulators of membrane voltage and as novel pharmacological targets for modulation of vascular contractility. In this issue of British Journal of Pharmacology, Ng et al., extend the findings from rodent models to the human vasculature and establish that KCNQ channels also regulate constriction of human arteries. The findings have important implications for the use of pharmacological KCNQ channel modulators to treat human diseases.
Collapse
Affiliation(s)
- Bharath K Mani
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA
| | | |
Collapse
|
89
|
Ng FL, Davis AJ, Jepps TA, Harhun MI, Yeung SY, Wan A, Reddy M, Melville D, Nardi A, Khong TK, Greenwood IA. Expression and function of the K+ channel KCNQ genes in human arteries. Br J Pharmacol 2011; 162:42-53. [PMID: 20840535 DOI: 10.1111/j.1476-5381.2010.01027.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE KCNQ-encoded voltage-gated potassium channels (K(v) 7) have recently been identified as important anti-constrictor elements in rodent blood vessels but the role of these channels and the effects of their modulation in human arteries remain unknown. Here, we have assessed KCNQ gene expression and function in human arteries ex vivo. EXPERIMENTAL APPROACH Fifty arteries (41 from visceral adipose tissue, 9 mesenteric arteries) were obtained from subjects undergoing elective surgery. Quantitative RT-PCR experiments using primers specific for all known KCNQ genes and immunohistochemsitry were used to show K(v) 7 channel expression. Wire myography and single cell electrophysiology assessed the function of these channels. KEY RESULTS KCNQ4 was expressed in all arteries assessed, with variable contributions from KCNQ1, 3 and 5. KCNQ2 was not detected. K(v) 7 channel isoform-dependent staining was revealed in the smooth muscle layer. In functional studies, the K(v) 7 channel blockers, XE991 and linopirdine increased isometric tension and inhibited K(+) currents. In contrast, the K(v) 7.1-specific blocker chromanol 293B did not affect vascular tone. Two K(v) 7 channel activators, retigabine and acrylamide S-1, relaxed preconstricted arteries, actions reversed by XE991. K(v) 7 channel activators also suppressed spontaneous contractile activity in seven arteries, reversible by XE991. CONCLUSIONS AND IMPLICATIONS This is the first study to demonstrate not only the presence of KCNQ gene products in human arteries but also their contribution to vascular tone ex vivo. LINKED ARTICLE This article is commented on by Mani and Byron, pp. 38-41 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2010.01065.x.
Collapse
Affiliation(s)
- Fu Liang Ng
- Biomedical Sciences, Ion Channels and Cell Signalling, St. George's, University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Kv7 (KCNQ) channel openers induce hypothermia in the mouse. Neurosci Lett 2011; 488:178-82. [DOI: 10.1016/j.neulet.2010.11.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/02/2010] [Accepted: 11/09/2010] [Indexed: 11/18/2022]
|
91
|
Abstract
BACKGROUND Perivascular adipose tissue secretes an adipocyte-derived relaxing factor (ADRF) that opens voltage-dependent K (Kv) channels in peripheral arteries. We studied the role of KCNQ-type Kv channels and tested the hypothesis that hydrogen sulfide (H2S) could be an ADRF. METHODS We performed isometric contraction studies on systemic arteries of rats and mice. RESULTS In mesenteric arteries and aortas without perivascular adipose tissue, the KCNQ channel openers retigabine, VRX0530727, VRX0621238, and VRX0621688 produced concentration-dependent vasorelaxation; VRX0621688 was the most potent vasodilator. The KCNQ inhibitor XE991 (30 micromol/l) blocked the effects of both the drugs and ADRF. Inhibitors of cystathionine gamma lyase (CSE) beta-cyano-L-alanine (BCA, 5 mmol/l) and 4-propargyl glycine (PPG, 10 mmol/l) also blocked the relaxations. CSE is expressed in perivascular adipose tissue and endogenously generates H2S. The H2S donor NaHS produced concentration-dependent vasorelaxation, which was also blocked by XE991. The vasodilatory capacities of retigabine, VRX0530727, VRX0621238, and VRX0621688 were preserved following inhibition of H2S generation in perivascular fat. CONCLUSION We suggest that KCNQ channel opening is a powerful mechanism to produce vasorelaxation of systemic arteries in rats and mice. Furthermore, KCNQ channels play a major role in the paracrine control of vascular tone by perivascular adipose tissue, which is at least in part mediated or modulated by H2S. In conditions of reduced H2S release from perivascular adipose tissue, these paracrine effects can be mimicked by synthetic KCNQ channel openers.
Collapse
|
92
|
Brueggemann LI, Mackie AR, Martin JL, Cribbs LL, Byron KL. Diclofenac distinguishes among homomeric and heteromeric potassium channels composed of KCNQ4 and KCNQ5 subunits. Mol Pharmacol 2010; 79:10-23. [PMID: 20876743 DOI: 10.1124/mol.110.067496] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
KCNQ4 and KCNQ5 potassium channel subunits are expressed in vascular smooth muscle cells, although it remains uncertain how these subunits assemble to form functional channels. Using patch-clamp techniques, we compared the electrophysiological characteristics and effects of diclofenac, a known KCNQ channel activator, on human KCNQ4 and KCNQ5 channels expressed individually or together in A7r5 rat aortic smooth muscle cells. The conductance curves of the overexpressed channels were fitted by a single Boltzmann function in each case (V(0.5) values: -31, -44, and -38 mV for KCNQ4, KCNQ5, and KCNQ4/5, respectively). Diclofenac (100 μM) inhibited KCNQ5 channels, reducing maximum conductance by 53%, but increased maximum conductance of KCNQ4 channels by 38%. The opposite effects of diclofenac on KCNQ4 and KCNQ5 could not be attributed to the presence of a basic residue (lysine) in the voltage-sensing domain of KCNQ5, because mutation of this residue to neutral glycine (the residue present in KCNQ4) resulted in a more effective block of the channel. Differences in deactivation rates and distinct voltage-dependent effects of diclofenac on channel activation and deactivation observed with each of the subunit combinations (KCNQ4, KCNQ5, and KCNQ4/5) were used as diagnostic tools to evaluate native KCNQ currents in vascular smooth muscle cells. A7r5 cells express only KCNQ5 channels endogenously, and their responses to diclofenac closely resembled those of the overexpressed KCNQ5 currents. In contrast, mesenteric artery myocytes, which express both KCNQ4 and KCNQ5 channels, displayed whole-cell KCNQ currents with properties and diclofenac responses characteristic of overexpressed heteromeric KCNQ4/5 channels.
Collapse
Affiliation(s)
- Lioubov I Brueggemann
- Department of Molecular Pharmacology and Therapeutics, Loyola University Medical Center,Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
93
|
Zhong XZ, Harhun MI, Olesen SP, Ohya S, Moffatt JD, Cole WC, Greenwood IA. Participation of KCNQ (Kv7) potassium channels in myogenic control of cerebral arterial diameter. J Physiol 2010; 588:3277-93. [PMID: 20624791 PMCID: PMC2976022 DOI: 10.1113/jphysiol.2010.192823] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 07/09/2010] [Indexed: 01/11/2023] Open
Abstract
KCNQ gene expression was previously shown in various rodent blood vessels, where the products of KCNQ4 and KCNQ5, Kv7.4 and Kv7.5 potassium channel subunits, respectively, have an influence on vascular reactivity. The aim of this study was to determine if small cerebral resistance arteries of the rat express KCNQ genes and whether Kv7 channels participate in the regulation of myogenic control of diameter. Quantitative reverse transcription polymerase chain reaction (QPCR) was undertaken using RNA isolated from rat middle cerebral arteries (RMCAs) and immunocytochemistry was performed using Kv7 subunit-specific antibodies and freshly isolated RMCA myocytes. KCNQ4 message was more abundant than KCNQ5 = KCNQ1, but KCNQ2 and KCNQ3 message levels were negligible. Kv7.1, Kv7.4 and Kv7.5 immunoreactivity was present at the sarcolemma of freshly isolated RMCA myocytes. Linopirdine (1 microm) partially depressed, whereas the Kv7 activator S-1 (3 and/or 20 microm) enhanced whole-cell Kv7.4 (in HEK 293 cells), as well as native RMCA myocyte Kv current amplitude. The effects of S-1 were voltage-dependent, with progressive loss of stimulation at potentials of >15 mV. At the concentrations employed linopirdine and S-1 did not alter currents due to recombinant Kv1.2/Kv1.5 or Kv2.1/Kv9.3 channels (in HEK 293 cells) that are also expressed by RMCA myocytes. In contrast, another widely used Kv7 blocker, XE991 (10 microm), significantly attenuated native Kv current and also reduced Kv1.2/Kv1.5 and Kv2.1/Kv9.3 currents. Pressurized arterial myography was performed using RMCAs exposed to intravascular pressures of 10-100 mmHg. Linopirdine (1 microm) enhanced the myogenic response at 20 mmHg, whereas the activation of Kv7 channels with S-1 (20 microm) inhibited myogenic constriction at >20 mmHg and reversed the increased myogenic response produced by suppression of Kv2-containing channels with 30 nm stromatoxin (ScTx1). These data reveal a novel contribution of KCNQ gene products to the regulation of myogenic control of cerebral arterial diameter and suggest that Kv7 channel activating drugs may be appropriate candidates for the development of an effective therapy to ameliorate cerebral vasospasm.
Collapse
Affiliation(s)
- Xi Zoë Zhong
- Ion Channels and Cell Signaling Centre, Division of Basic Medical Sciences, St George's University of London, London SW17 0RE, UK
| | | | | | | | | | | | | |
Collapse
|
94
|
Functional effects of the KCNQ modulators retigabine and XE991 in the rat urinary bladder. Eur J Pharmacol 2010; 638:121-7. [PMID: 20385123 DOI: 10.1016/j.ejphar.2010.03.050] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 02/16/2010] [Accepted: 03/24/2010] [Indexed: 01/15/2023]
Abstract
The anticonvulsant retigabine has previously been reported to inhibit bladder overactivity in rats in vivo but the mechanism and site of action are not known. In the present study we investigated the effect of retigabine in isolated rat bladder tissue. Bladders from Sprague-Dawley rats were cut transversally into rings and mounted on an isometric myograph. The average tension, the amplitude and frequency of bladder muscle twitches were measured. The bladder tissue was stimulated with carbachol, KCl (5, 10 and 60mM), and by electric field stimulation. Dose-response curves were obtained with increasing concentrations of the KCNQ((2-5)) selective positive modulator, retigabine or with the KCNQ((1-5)) negative modulator XE991. Retigabine experiments were repeated in the presence of 10 microM XE991. Retigabine reduced both the contractility and the overall tonus of bladder tissue independent of the mode of stimulation with EC(50) values ranging from 3.3 microM (20mM KCl) to 8.3 microM (0.2 microM carbachol). In support of a KCNQ-specific effect, retigabine had only weak effects after 60mM KCl pre treatment and all retigabine effects could be reversed by XE991. XE991 increased both the amplitude and mean tension of the bladder but was more potent at increasing the number rather than the size of the stimulated twitches. In conclusion, this study demonstrates an efficacious KCNQ dependent effect of retigabine and XE991 on rat bladder contractility.
Collapse
|
95
|
Gurney AM, Joshi S, Manoury B. KCNQ potassium channels: new targets for pulmonary vasodilator drugs? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:405-17. [PMID: 20204745 DOI: 10.1007/978-1-60761-500-2_26] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Smooth muscle cells regulate the diameter of pulmonary arteries and the resistance to blood flow in the pulmonary circulation. These cells are normally relaxed to maintain low intrinsic vessel tone, but are contracted in pulmonary arterial hypertension (PAH). Potassium channels in the smooth muscle cell help to maintain low tone by polarising the membrane and preventing Ca(2+) influx through voltage-operated Ca(2+) channels. There is a loss of K(+) channel activity in PAH, so drugs that open K(+) channels are predicted to have a beneficial effect, provided their action can be restricted to the pulmonary circulation. Here we review the myriad of K(+) channels that are expressed in pulmonary arteries and suggest the roles that each might play in regulating pulmonary artery tone. We conclude that members of the KCNQ family of K(+) channels, the most recent K(+) channels to be discovered in pulmonary artery, may be a useful therapeutic target for the treatment of PAH. KCNQ channels appear to be preferentially expressed in pulmonary arteries and drugs that modulate their activity have potent effects on pulmonary artery tone.
Collapse
Affiliation(s)
- Alison M Gurney
- Faculty of Life Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK, Alison.
| | | | | |
Collapse
|
96
|
Brueggemann LI, Mani BK, Mackie AR, Cribbs LL, Byron KL. Novel Actions of Nonsteroidal Anti-Inflammatory Drugs on Vascular Ion Channels: Accounting for Cardiovascular Side Effects and Identifying New Therapeutic Applications. MOLECULAR AND CELLULAR PHARMACOLOGY 2010; 2:15-19. [PMID: 20689646 PMCID: PMC2915785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used medications for the treatment of both acute and chronic pain. Selective cyclooxygenase-2 (COX-2) inhibitors, such as celecoxib (Celebrex(®)), rofecoxib (Vioxx(®)), and diclofenac, have been among the most widely prescribed NSAIDs because they prevent the generation of prostaglandins involved in inflammation and pain, but avoid some of the gastrointestinal complications associated with less selective COX-1/COX-2 inhibitors. In 2004, rofecoxib (Vioxx(®)) was voluntarily withdrawn from the market because of adverse cardiovascular side effects. This led to an explosion of research into the cardiovascular effects of the 'coxibs', which revealed differential cardiovascular risk profiles among the members of this drug class. The differential risk profiles may relate to the tendency of some of the drugs to elevate blood pressure (BP). An important component of BP regulation is dependent on the contractile state of vascular smooth muscle cells (VSMCs), which is controlled to a large extent by the activities of KCNQ (Kv7 family) potassium channels and L-type calcium channels. Our recently published data indicate that celecoxib, but not rofecoxib or diclofenac, at therapeutically relevant concentrations, acts as a Kv7 potassium channel activator and a calcium channel blocker, causing relaxation of VSMCs and decreasing vascular tone. These vasorelaxant ion channel effects may account for the differential cardiovascular risk profiles among the different COX-2 inhibitors. We further speculate that these properties may be exploited for therapeutic benefit in the treatment of cardiovascular diseases or other medical conditions.
Collapse
Affiliation(s)
- Lioubov I Brueggemann
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, Illinois
| | | | | | | | | |
Collapse
|
97
|
Manoury B, Etheridge SL, Reid J, Gurney AM. Organ culture mimics the effects of hypoxia on membrane potential, K(+) channels and vessel tone in pulmonary artery. Br J Pharmacol 2009; 158:848-61. [PMID: 19694728 DOI: 10.1111/j.1476-5381.2009.00353.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Blood vessel culture is gaining interest for use with transfection-based techniques, but alters the contractile properties of the vessels. The present study tested the effects of culture on the intrinsic tone of rat pulmonary arteries (PAs) and examined the function and expression of K(+) channels regulating the resting membrane potential (E(m)) and tone of pulmonary artery smooth muscle cells (PASMCs). EXPERIMENTAL APPROACH Rat intrapulmonary arteries were isolated and cultured under standard and modified conditions. Contractile responses of fresh and cultured PA were compared using vessel myograph. Electrophysiology experiments on isolated PASMCs used the patch-clamp technique. K(+) channel expression was quantified using reverse transcription and real-time PCR. KEY RESULTS After 4 days in culture vessels contracted to phenylephrine, but relaxation to carbachol was significantly impaired. Contractile responses to 10 mM KCl, 4-aminopyridine and tetraethylammonium increased, and vessels developed an uncharacteristic relaxation response to Ca(2+)-free solution, nifedipine and levcromakalim. PASMCs from cultured vessels were depolarized and K(+) currents reduced, in association with down-regulation of K(v)1.5, K(v)2.1 and TWIK-related acid-sensitive K(+) channel-1 mRNA. These changes were partially reversed by increased oxygenation of the culture medium or removing the endothelium before culture. CONCLUSIONS AND IMPLICATIONS Culture of PA for 3-4 days induced loss of functional K(+) channels, depolarization of PASMCs, Ca(2+) influx, intrinsic tone and spontaneous constrictions, similar to the effects of chronic hypoxia. This limits the use of cultured vessels for studying excitation-contraction coupling, although oxygenating the culture medium and removing the endothelium can help to retain normal smooth muscle function.
Collapse
|
98
|
Morecroft I, Murray A, Nilsen M, Gurney AM, MacLean MR. Treatment with the Kv7 potassium channel activator flupirtine is beneficial in two independent mouse models of pulmonary hypertension. Br J Pharmacol 2009; 157:1241-9. [PMID: 19508393 PMCID: PMC2743843 DOI: 10.1111/j.1476-5381.2009.00283.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/16/2009] [Accepted: 02/23/2009] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Voltage-gated potassium (K(v)) channels contribute to resting membrane potential in pulmonary artery smooth muscle cells and are down regulated in patients with pulmonary arterial hypertension (PAH) and a contribution from K(v)7 channels has been recently proposed. We investigated the effect of the K(v)7 channel activator, flupirtine, on PAH in two independent mouse models: PAH induced by hypoxia and spontaneous PAH in mice over-expressing the 5-HT transporter (SERT(+) mice). EXPERIMENTAL APPROACH Right ventricular pressure was assessed in vivo in mice chronically treated with flupirtine (30 mg.kg(-1).day(-1)). In separate in vitro experiments, pulmonary arteries from untreated mice were mounted in a wire myograph. Relaxations to acute administration of flupirtine and contractions to K(v) channel blocking drugs, including the K(v)7 channel blocker linopirdine, were measured. KEY RESULTS In wild-type (WT) mice, hypoxia increased right ventricular pressure, pulmonary vascular remodelling and right ventricular hypertrophy. These effects were attenuated by flupirtine, which also attenuated these indices of PAH in SERT(+) mice. In the in vitro experiments, flupirtine induced a potent relaxant response in arteries from untreated WT and SERT(+) mice. The relaxation was fully reversed by linopirdine, which potently contracted mouse pulmonary arteries while other K(v) channel blockers did not. CONCLUSIONS AND IMPLICATIONS Flupirtine significantly attenuated development of chronic hypoxia-induced PAH in mice and reversed established PAH in SERT(+) mice, apparently via K(v)7 channel activation. These results provide the first direct evidence that drugs activating K(v)7 channels may be of benefit in the treatment of PAH with different aetiologies.
Collapse
MESH Headings
- Aminopyridines/therapeutic use
- Animals
- Disease Models, Animal
- Female
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/physiopathology
- Hypoxia/complications
- In Vitro Techniques
- Mice
- Muscle Contraction/drug effects
- Muscle Relaxation/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Potassium Channels, Voltage-Gated/agonists
- Potassium Channels, Voltage-Gated/antagonists & inhibitors
- Pulmonary Artery/drug effects
- Pulmonary Artery/physiopathology
- Serotonin Plasma Membrane Transport Proteins/genetics
Collapse
Affiliation(s)
- I Morecroft
- Integrative and Systems Biology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK.
| | | | | | | | | |
Collapse
|
99
|
Blockade of the IKs potassium channel: An overlooked cardiovascular liability in drug safety screening? J Pharmacol Toxicol Methods 2009; 60:1-10. [DOI: 10.1016/j.vascn.2009.04.197] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 04/25/2009] [Indexed: 12/25/2022]
|