1
|
Studley WR, Lamanna E, Martin KA, Nold-Petry CA, Royce SG, Woodman OL, Ritchie RH, Qin CX, Bourke JE. The small-molecule formyl peptide receptor biased agonist, compound 17b, is a vasodilator and anti-inflammatory in mouse precision-cut lung slices. Br J Pharmacol 2024; 181:2287-2301. [PMID: 37658546 DOI: 10.1111/bph.16231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary arterial hypertension (PAH), a rare fatal disorder characterised by inflammation, vascular remodelling and vasoconstriction. Current vasodilator therapies reduce pulmonary arterial pressure but not mortality. The G-protein coupled formyl peptide receptors (FPRs) mediates vasodilatation and resolution of inflammation, actions possibly beneficial in PAH. We investigated dilator and anti-inflammatory effects of the FPR biased agonist compound 17b in pulmonary vasculature using mouse precision-cut lung slices (PCLS). EXPERIMENTAL APPROACH PCLS from 8-week-old male and female C57BL/6 mice, intrapulmonary arteries were pre-contracted with 5-HT for concentration-response curves to compound 17b and 43, and standard-of-care drugs, sildenafil, iloprost and riociguat. Compound 17b-mediated relaxation was assessed with FPR antagonists or inhibitors and in PCLS treated with TNF-α or LPS. Cytokine release from TNF-α- or LPS-treated PCLS ± compound 17b was measured. KEY RESULTS Compound 17b elicited concentration-dependent vasodilation, with potencies of iloprost > compound 17b = riociguat > compound 43 = sildenafil. Compound 17b was inhibited by the FPR1 antagonist cyclosporin H but not by soluble guanylate cyclase, nitric oxide synthase or cyclooxygenase inhibitors. Under inflammatory conditions, the efficacy and potency of compound 17b were maintained, while iloprost and sildenafil were less effective. Additionally, compound 17b inhibited secretion of PAH-relevant cytokines via FPR2. CONCLUSIONS AND IMPLICATIONS Vasodilation to compound 17b but not standard-of-care vasodilators, is maintained under inflammatory conditions, with additional inhibition of PAH-relevant cytokine release. This provides the first evidence that targeting FPR, with biased agonist, simultaneously targets vascular function and inflammation, supporting the development of FPR-based pharmacotherapy to treat PAH. LINKED ARTICLES This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- William R Studley
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Emma Lamanna
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Katherine A Martin
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Claudia A Nold-Petry
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Simon G Royce
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Owen L Woodman
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rebecca H Ritchie
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Cheng Xue Qin
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Stott JB, Greenwood IA. G protein βγ regulation of KCNQ-encoded voltage-dependent K channels. Front Physiol 2024; 15:1382904. [PMID: 38655029 PMCID: PMC11035767 DOI: 10.3389/fphys.2024.1382904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The KCNQ family is comprised of five genes and the expression products form voltage-gated potassium channels (Kv7.1-7.5) that have a major impact upon cellular physiology in many cell types. Each functional Kv7 channel forms as a tetramer that often associates with proteins encoded by the KCNE gene family (KCNE1-5) and is critically reliant upon binding of phosphatidylinositol bisphosphate (PIP2) and calmodulin. Other modulators like A-kinase anchoring proteins, ubiquitin ligases and Ca-calmodulin kinase II alter Kv7 channel function and trafficking in an isoform specific manner. It has now been identified that for Kv7.4, G protein βγ subunits (Gβγ) can be added to the list of key regulators and is paramount for channel activity. This article provides an overview of this nascent field of research, highlighting themes and directions for future study.
Collapse
Affiliation(s)
| | - Iain A. Greenwood
- Vascular Biology Research Group, Institute of Molecular and Clinical Sciences, St George’s University of London, London, United Kingdom
| |
Collapse
|
3
|
Zhang L, Sunchen S, Lu C, Xu F, Dong H. Zinc-sensing receptor activation induces endothelium-dependent hyperpolarization-mediated vasorelaxation of arterioles. Biochem Pharmacol 2024; 219:115961. [PMID: 38049010 DOI: 10.1016/j.bcp.2023.115961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND The micronutrient zinc (Zn2+) is critical for cell function as intracellular signaling and endogenous ligand for Zn2+ sensing receptor (ZnR). Although cytosolic Zn2+ (cyt) signaling in the vascular system was studied previously, role of the ZnR has not been explored in vascular physiology. METHODS ZnR-mediated relaxation response of human submucosal arterioles and the mesenteric arterioles from wide-type (WT), ZnR-/- and TRPV4-/- mice were determined by a Mulvany-style wire myograph. The perfused vessel density (PVD) of mouse mesenteric arterioles was also measured in in vivo study. The expression of ZnR in arterioles and vascular endothelial cells (VEC) were examined by immunofluorescence staining, and its function was characterized in VEC by Ca2+ imaging and patch clamp study. RESULTS ZnR expression was detected on human submucosal arterioles, murine mesenteric arterioles and VEC but not in ZnR-/- mice. ZnR activation predominately induced endothelium-dependent hyperpolarization (EDH)-mediated vasorelaxation of arterioles in vitro and in vivo via Ca2+ signaling, which is totally different from endothelium-dependent vasorelaxation via Zn2+ (cyt) signaling reported previously. Furthermore, ZnR-induced vasorelaxation via EDH was significantly impaired in ZnR-/- and TRPV4-/- mice. Mechanistically, ZnR induced endothelium-dependent vasorelaxation predominately via PLC/IP3/IP3R and TRPV4/SOCE. The role of ZnR in regulating Ca2+ signaling and ion channels on VEC was verified by Ca2+ imaging and patch clamp techniques. CONCLUSION ZnR activation induces endothelium-dependent vasorelaxation of resistance vessels predominately via TRPV4/Ca2+/EDH pathway. We therefore not only provide new insights into physiological role of ZnR in vascular system but also may pave a potential pathway for developing Zn2+-based treatments for vascular disease.
Collapse
Affiliation(s)
- Luyun Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400037, China; Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Sijin Sunchen
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400037, China.
| | - Hui Dong
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400037, China; Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
4
|
Baldwin SN, Jepps TA, Greenwood IA. Cycling matters: Sex hormone regulation of vascular potassium channels. Channels (Austin) 2023; 17:2217637. [PMID: 37243715 PMCID: PMC10228406 DOI: 10.1080/19336950.2023.2217637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/07/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023] Open
Abstract
Sex hormones and the reproductive cycle (estrus in rodents and menstrual in humans) have a known impact on arterial function. In spite of this, sex hormones and the estrus/menstrual cycle are often neglected experimental factors in vascular basic preclinical scientific research. Recent research by our own laboratory indicates that cyclical changes in serum concentrations of sex -hormones across the rat estrus cycle, primary estradiol, have significant consequences for the subcellular trafficking and function of KV. Vascular potassium channels, including KV, are essential components of vascular reactivity. Our study represents a small part of a growing field of literature aimed at determining the role of sex hormones in regulating arterial ion channel function. This review covers key findings describing the current understanding of sex hormone regulation of vascular potassium channels, with a focus on KV channels. Further, we highlight areas of research where the estrus cycle should be considered in future studies to determine the consequences of physiological oscillations in concentrations of sex hormones on vascular potassium channel function.
Collapse
Affiliation(s)
- Samuel N Baldwin
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George’s University of London, London, UK
| |
Collapse
|
5
|
Shan X, Gegentuya, Wang J, Feng H, Zhang Z, Zheng Q, Zhang Q, Yang K, Wang J, Xu L. Aloperine protects pulmonary hypertension via triggering PPARγ signaling and inhibiting calcium regulatory pathway in pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2023; 325:C1058-C1072. [PMID: 37661916 DOI: 10.1152/ajpcell.00286.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Previous studies have reported the beneficial role of Aloperine (ALO), an active vasodilator purified from the seeds and leaves of the herbal plant Sophora alopecuroides L., on experimental pulmonary hypertension (PH); however, detailed mechanisms remain unclear. In this study, monocrotaline-induced PH (MCT-PH) rat model and primarily cultured rat distal pulmonary arterial smooth muscle cells (PASMCs) were used to investigate the mechanisms of ALO on experimental PH, pulmonary vascular remodeling, and excessive proliferation of PASMCs. Results showed that first, ALO significantly prevented the disease development of MCT-PH by inhibiting right ventricular systolic pressure (RVSP) and right ventricular hypertrophy indexed by the Fulton Index, normalizing the pulmonary arterials (PAs) remodeling and improving the right ventricular function indexed by transthoracic echocardiography. ALO inhibited the excessive proliferation of both PAs and PASMCs. Then, isometric tension measurements showed vasodilation of ALO on precontracted PAs isolated from both control and MCT-PH rats via activating the KCNQ channel, which was blocked by specific KCNQ potassium channel inhibitor linopirdine. Moreover, by using immunofluorescence staining and nuclear/cytosol fractionation, we further observed that ALO significantly enhanced the PPARγ nuclear translocation and activation in PASMCs. Transcriptome analyses also revealed activated PPARγ signaling and suppressed calcium regulatory pathway in lungs from MCT-PH rats treated with ALO. In summary, ALO could attenuate MCT-PH through both transient vasodilation of PAs and chronic activation of PPARγ signaling pathway, which exerted antiproliferative roles on PASMCs and remodeled PAs.NEW & NOTEWORTHY Aloperine attenuates monocrotaline-induced pulmonary hypertension (MCT-PH) in rats by inhibiting the pulmonary vascular remodeling and proliferation of pulmonary arterial smooth muscle cells (PASMCs). In mechanism, Aloperine not only exerts a transient KCNQ-dependent vasodilation in precontracted pulmonary arteries (PAs) from both control and MCT-PH rats but also activates PPARγ nuclear translocation and signaling transduction in PASMCs, which chronically inhibits the calcium regulatory pathway and proliferation of PASMCs.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Calcium/metabolism
- Calcium Signaling/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- KCNQ Potassium Channels/metabolism
- KCNQ Potassium Channels/genetics
- Monocrotaline/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Piperidines/pharmacology
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Quinolizidines/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Xiaoqian Shan
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Gegentuya
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Jing Wang
- Department of Scientific Research, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zizhou Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Qiuyu Zheng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qing Zhang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
6
|
Ehlers TS, van der Horst J, Møller S, Piil PK, Gliemann L, Aalkjaer C, Jepps TA, Hellsten Y. Colchicine enhances β adrenoceptor-mediated vasodilation in men with essential hypertension. Br J Clin Pharmacol 2023; 89:2179-2189. [PMID: 36764326 DOI: 10.1111/bcp.15688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 02/12/2023] Open
Abstract
AIMS The aim of this study is to examine whether colchicine improves β adrenoceptor-mediated vasodilation in humans by conducting a double-blinded, placebo-controlled intervention study. Colchicine treatment has known beneficial effects on cardiovascular health and reduces the incidence of cardiovascular disease. Studies in isolated rodent arteries have shown that colchicine can enhance β adrenoceptor-mediated vasodilation, but this has not been determined in humans. METHODS Middle-aged men with essential hypertension were randomly assigned firstly to acute treatment with either 0.5 mg colchicine (n = 19) or placebo (n = 12). They were subsequently re-randomized for 3 weeks of treatment with either colchicine 0.5 mg twice daily (n = 16) or placebo (n = 15) followed by a washout period of 48-72 h. The vasodilator responses to isoprenaline, acetylcholine and sodium nitroprusside were determined as well as arterial pressure, arterial compliance and plasma inflammatory markers. RESULTS Acute colchicine treatment increased isoprenaline (by 38% for the highest dose) as well as sodium nitroprusside (by 29% main effect) -induced vasodilation but had no effect on the response to acetylcholine. The 3-week colchicine treatment followed by a washout period did not induce an accumulated or sustained effect on the β adrenoceptor response, and there was no effect on arterial pressure, arterial compliance or the level of measured inflammatory markers. CONCLUSION Colchicine acutely enhances β adrenoceptor- and nitric oxide-mediated changes in vascular conductance in humans, supporting that the mechanism previously demonstrated in rodents, translates to humans. The results provide novel translational evidence for a transient enhancing effect of colchicine on β adrenoceptor-mediated vasodilation in humans with essential hypertension. CONDENSED ABSTRACT Preclinical studies in isolated rodent arteries have shown that colchicine can enhance β adrenoceptor-mediated vasodilation. Here we show that this effect of colchicine can be translated to humans. Acute colchicine treatment was found to increase both isoprenaline- and sodium nitroprusside-induced vasodilation. The study provides the first translational evidence for a transient β adrenoceptor-mediated vasodilatory effect of colchicine in humans. The finding of an acute effect suggests that it may be clinically important to maintain an adequate bioavailability of colchicine.
Collapse
Affiliation(s)
- Thomas S Ehlers
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Jennifer van der Horst
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Møller
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Peter K Piil
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Lasse Gliemann
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Christian Aalkjaer
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Baldwin SN, Forrester EA, Homer NZM, Andrew R, Barrese V, Stott JB, Isakson BE, Albert AP, Greenwood IA. Marked oestrous cycle-dependent regulation of rat arterial K V 7.4 channels driven by GPER1. Br J Pharmacol 2023; 180:174-193. [PMID: 36085551 PMCID: PMC10091994 DOI: 10.1111/bph.15947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 06/21/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Kcnq-encoded KV 7 channels (termed KV 7.1-5) regulate vascular smooth muscle cell (VSMC) contractility at rest and as targets of receptor-mediated responses. However, the current data are mostly derived from males. Considering the known effects of sex, the oestrous cycle and sex hormones on vascular reactivity, here we have characterised the molecular and functional properties of KV 7 channels from renal and mesenteric arteries from female Wistar rats separated into di-oestrus and met-oestrus (F-D/M) and pro-oestrus and oestrus (F-P/E). EXPERIMENTAL APPROACH RT-qPCR, immunocytochemistry, proximity ligation assay and wire myography were performed in renal and mesenteric arteries. Circulating sex hormone concentrations were determined by liquid chromatography-tandem mass spectrometry. Whole-cell electrophysiology was undertaken on cells expressing KV 7.4 channels in association with G-protein-coupled oestrogen receptor 1 (GPER1). KEY RESULTS The KV 7.2-5 activators S-1 and ML213 and the pan-KV 7 inhibitor linopirdine were more effective in arteries from F-D/M compared with F-P/E animals. In VSMCs isolated from F-P/E rats, exploratory evidence indicates reduced membrane abundance of KV 7.4 but not KV 7.1, KV 7.5 and Kcne4 when compared with cells from F-D/M. Plasma oestradiol was higher in F-P/E compared with F-D/M, and progesterone showed the converse pattern. Oestradiol/GPER1 agonist G-1 diminished KV 7.4 encoded currents and ML213 relaxations and reduced the membrane abundance of KV 7.4 and interaction between KV 7.4 and heat shock protein 90 (HSP90), in arteries from F-D/M but not F-P/E. CONCLUSIONS AND IMPLICATIONS GPER1 signalling decreased KV 7.4 membrane abundance in conjunction with diminished interaction with HSP90, giving rise to a 'pro-contractile state'.
Collapse
Affiliation(s)
- Samuel N. Baldwin
- Vascular Biology Research Centre, Institute of Molecular and Clinical SciencesSt George's University of LondonLondonUK
| | - Elizabeth A. Forrester
- Vascular Biology Research Centre, Institute of Molecular and Clinical SciencesSt George's University of LondonLondonUK
| | - Natalie Z. M. Homer
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Ruth Andrew
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
- BHF Centre for Cardiovascular Science, Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Vincenzo Barrese
- Department of Neuroscience, Reproductive Sciences and DentistryUniversity of Naples Federico IINaplesItaly
| | - Jennifer B. Stott
- Vascular Biology Research Centre, Institute of Molecular and Clinical SciencesSt George's University of LondonLondonUK
| | - Brant E. Isakson
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research CentreUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Anthony P. Albert
- Vascular Biology Research Centre, Institute of Molecular and Clinical SciencesSt George's University of LondonLondonUK
| | - Iain A. Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical SciencesSt George's University of LondonLondonUK
| |
Collapse
|
8
|
Redel-Traub G, Sampson KJ, Kass RS, Bohnen MS. Potassium Channels as Therapeutic Targets in Pulmonary Arterial Hypertension. Biomolecules 2022; 12:1341. [PMID: 36291551 PMCID: PMC9599705 DOI: 10.3390/biom12101341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 12/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease with high morbidity and mortality. Deleterious remodeling in the pulmonary arterial system leads to irreversible arterial constriction and elevated pulmonary arterial pressures, right heart failure, and eventually death. The difficulty in treating PAH stems in part from the complex nature of disease pathogenesis, with several signaling compounds known to be involved (e.g., endothelin-1, prostacyclins) which are indeed targets of PAH therapy. Over the last decade, potassium channelopathies were established as novel causes of PAH. More specifically, loss-of-function mutations in the KCNK3 gene that encodes the two-pore-domain potassium channel KCNK3 (or TASK-1) and loss-of-function mutations in the ABCC8 gene that encodes a key subunit, SUR1, of the ATP-sensitive potassium channel (KATP) were established as the first two potassium channelopathies in human cohorts with pulmonary arterial hypertension. Moreover, voltage-gated potassium channels (Kv) represent a third family of potassium channels with genetic changes observed in association with PAH. While other ion channel genes have since been reported in association with PAH, this review focuses on KCNK3, KATP, and Kv potassium channels as promising therapeutic targets in PAH, with recent experimental pharmacologic discoveries significantly advancing the field.
Collapse
Affiliation(s)
- Gabriel Redel-Traub
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kevin J. Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Robert S. Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael S. Bohnen
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
9
|
Kv7 Channels in Cyclic-Nucleotide Dependent Relaxation of Rat Intra-Pulmonary Artery. Biomolecules 2022; 12:biom12030429. [PMID: 35327621 PMCID: PMC8946781 DOI: 10.3390/biom12030429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension is treated with drugs that stimulate cGMP or cAMP signalling. Both nucleotides can activate Kv7 channels, leading to smooth muscle hyperpolarisation, reduced Ca2+ influx and relaxation. Kv7 activation by cGMP contributes to the pulmonary vasodilator action of nitric oxide, but its contribution when dilation is evoked by the atrial natriuretic peptide (ANP) sensitive guanylate cyclase, or cAMP, is unknown. Small vessel myography was used to investigate the ability of Kv7 channel blockers to interfere with pulmonary artery relaxation when cyclic nucleotide pathways were stimulated in different ways. The pan-Kv7 blockers, linopirdine and XE991, caused substantial inhibition of relaxation evoked by NO donors and ANP, as well as endothelium-dependent dilators, the guanylate cyclase stimulator, riociguat, and the phosphodiesterase-5 inhibitor, sildenafil. Maximum relaxation was reduced without a change in sensitivity. The blockers had relatively little effect on cAMP-mediated relaxation evoked by forskolin, isoprenaline or treprostinil. The Kv7.1-selective blocker, HMR1556, had no effect on cGMP or cAMP-dependent relaxation. Western blot analysis demonstrated the presence of Kv7.1 and Kv7.4 proteins, while selective activators of Kv7.1 and Kv7.4 homomeric channels, but not Kv7.5, caused pulmonary artery relaxation. It is concluded that Kv7.4 channels contribute to endothelium-dependent dilation and the effects of drugs that act by stimulating cGMP, but not cAMP, signalling.
Collapse
|
10
|
Tettey A, Jiang Y, Li X, Li Y. Therapy for Pulmonary Arterial Hypertension: Glance on Nitric Oxide Pathway. Front Pharmacol 2021; 12:767002. [PMID: 34867394 PMCID: PMC8633825 DOI: 10.3389/fphar.2021.767002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with a resultant increase of the mean pulmonary arterial pressure, right ventricular hypertrophy and eventual death. Research in recent years has produced various therapeutic options for its clinical management but the high mortality even under treatment remains a big challenge attributed to the complex pathophysiology. Studies from clinical and non-clinical experiments have revealed that the nitric oxide (NO) pathway is one of the key pathways underlying the pathophysiology of PAH. Many of the essential drugs used in the management of PAH act on this pathway highlighting its significant role in PAH. Meanwhile, several novel compounds targeting on NO pathway exhibits great potential to become future therapy medications. Furthermore, the NO pathway is found to interact with other crucial pathways. Understanding such interactions could be helpful in the discovery of new drug that provide better clinical outcomes.
Collapse
Affiliation(s)
- Abraham Tettey
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| |
Collapse
|
11
|
Perez-Vizcaino F, Cogolludo A, Mondejar-Parreño G. Transcriptomic profile of cationic channels in human pulmonary arterial hypertension. Sci Rep 2021; 11:15829. [PMID: 34349187 PMCID: PMC8338963 DOI: 10.1038/s41598-021-95196-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/19/2021] [Indexed: 12/27/2022] Open
Abstract
The dysregulation of K+ channels is a hallmark of pulmonary arterial hypertension (PAH). Herein, the channelome was analyzed in lungs of patients with PAH in a public transcriptomic database. Sixty six (46%) mRNA encoding cationic channels were dysregulated in PAH with most of them downregulated (83%). The principal component analysis indicated that dysregulated cationic channel expression is a signature of the disease. Changes were very similar in idiopathic, connective tissue disease and congenital heart disease associated PAH. This analysis 1) is in agreement with the widely recognized pathophysiological role of TASK1 and KV1.5, 2) supports previous preliminary reports pointing to the dysregulation of several K+ channels including the downregulation of KV1.1, KV1.4, KV1.6, KV7.1, KV7.4, KV9.3 and TWIK2 and the upregulation of KCa1.1 and 3) points to other cationic channels dysregulated such as Kv7.3, TALK2, CaV1 and TRPV4 which might play a pathophysiological role in PAH. The significance of other changes found in Na+ and TRP channels remains to be investigated.
Collapse
Affiliation(s)
- Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology. School of Medicine, Universidad Complutense de Madrid, Madrid, Spain. .,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain. .,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain.
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology. School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Gema Mondejar-Parreño
- Department of Pharmacology and Toxicology. School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| |
Collapse
|
12
|
Beck L, Pinilla E, Arcanjo DDR, Hernanz R, Prat-Duran J, Petersen AG, Köhler R, Sheykhzade M, Comerma-Steffensen S, Simonsen U. Pirfenidone Is a Vasodilator: Involvement of K V7 Channels in the Effect on Endothelium-Dependent Vasodilatation in Type-2 Diabetic Mice. Front Pharmacol 2021; 11:619152. [PMID: 33643042 PMCID: PMC7906977 DOI: 10.3389/fphar.2020.619152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell dysfunction and fibrosis are associated with worsening of the prognosis in patients with cardiovascular disease. Pirfenidone has a direct antifibrotic effect, but vasodilatation may also contribute to the effects of pirfenidone. Therefore, in a first study we investigated the mechanisms involved in the relaxant effect of pirfenidone in rat intrapulmonary arteries and coronary arteries from normal mice. Then in a second study, we investigated whether pirfenidone restores endothelial function in the aorta and mesenteric arteries from diabetic animals. From 16–18-week old normal male C57BL/6 mice and normoglycemic (db/db+), and type 2 diabetic (db/db) male and female mice, arteries were mounted in microvascular isometric myographs for functional studies, and immunoblotting was performed. In rat pulmonary arteries and mouse coronary arteries, pirfenidone induced relaxations, which were inhibited in preparations without endothelium. In mouse coronary arteries, pirfenidone relaxation was inhibited in the presence of a nitric oxide (NO) synthase inhibitor, NG-nitro-l-arginine (L-NOARG), a blocker of large-conductance calcium-activated potassium channels (BKCa), iberiotoxin, and a blocker of KV7 channels, XE991. Patch clamp studies in vascular smooth muscle revealed pirfenidone increased iberiotoxin-sensitive current. In the aorta and mesenteric small arteries from diabetic db/db mice relaxations induced by the endothelium-dependent vasodilator, acetylcholine, were markedly reduced compared to db/db + mice. Pirfenidone enhanced the relaxations induced by acetylcholine in the aorta from diabetic male and female db/db mice. An opener of KV7 channels, flupirtine, had the same effect as pirfenidone. XE991 reduced the effect of pirfenidone and flupirtine and further reduced acetylcholine relaxations in the aorta. In the presence of iberiotoxin, pirfenidone still increased acetylcholine relaxation in aorta from db/db mice. Immunoblotting for KV7.4, KV7.5, and BKCa channel subunits were unaltered in aorta from db/db mice. Pirfenidone failed to improve acetylcholine relaxation in mesenteric arteries, and neither changed acetylcholine-induced transient decreases in blood pressure in db/db+ and db/db mice. In conclusion, pirfenidone vasodilates pulmonary and coronary arteries. In coronary arteries from normal mice, pirfenidone induces NO-dependent vasodilatation involving BKCa and KV7 channels. Pirfenidone improves endothelium-dependent vasodilatation in aorta from diabetic animals by a mechanism involving voltage-gated KV7 channels, a mechanism that may contribute to the antifibrotic effect of pirfenidone.
Collapse
Affiliation(s)
- Lilliana Beck
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Physiology, Faculty of Pharmacy, Universidad Complutense, Madrid, Spain
| | - Daniel Dias Rufino Arcanjo
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biophysics and Physiology, Laboratory of Functional and Molecular Studies in Physiopharmacology, Federal University of Piauí, Teresina, Brazil
| | - Raquel Hernanz
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Judit Prat-Duran
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Asbjørn Graver Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Ralf Köhler
- Aragón Agency for Research and Development (ARAID), Zaragoza, Spain
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biomedical Sciences/Animal Physiology, Faculty of Veterinary, Central University of Venezuela, Maracay, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Baldwin SN, Sandow SL, Mondéjar-Parreño G, Stott JB, Greenwood IA. K V7 Channel Expression and Function Within Rat Mesenteric Endothelial Cells. Front Physiol 2020; 11:598779. [PMID: 33364977 PMCID: PMC7750541 DOI: 10.3389/fphys.2020.598779] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/13/2020] [Indexed: 01/04/2023] Open
Abstract
Background and Purpose: Arterial diameter is dictated by the contractile state of the vascular smooth muscle cells (VSMCs), which is modulated by direct and indirect inputs from endothelial cells (ECs). Modulators of KCNQ-encoded kV7 channels have considerable impact on arterial diameter and these channels are known to be expressed in VSMCs but not yet defined in ECs. However, expression of kV7 channels in ECs would add an extra level of vascular control. This study aims to characterize the expression and function of KV7 channels within rat mesenteric artery ECs. Experimental Approach: In rat mesenteric artery, KCNQ transcript and KV7 channel protein expression were determined via RT-qPCR, immunocytochemistry, immunohistochemistry and immunoelectron microscopy. Wire myography was used to determine vascular reactivity. Key Results: KCNQ transcript was identified in isolated ECs and VSMCs. KV7.1, KV7.4 and KV7.5 protein expression was determined in both isolated EC and VSMC and in whole vessels. Removal of ECs attenuated vasorelaxation to two structurally different KV7.2-5 activators S-1 and ML213. KIR2 blockers ML133, and BaCl2 also attenuated S-1 or ML213-mediated vasorelaxation in an endothelium-dependent process. KV7 inhibition attenuated receptor-dependent nitric oxide (NO)-mediated vasorelaxation to carbachol, but had no impact on relaxation to the NO donor, SNP. Conclusion and Implications: In rat mesenteric artery ECs, KV7.4 and KV7.5 channels are expressed, functionally interact with endothelial KIR2.x channels and contribute to endogenous eNOS-mediated relaxation. This study identifies KV7 channels as novel functional channels within rat mesenteric ECs and suggests that these channels are involved in NO release from the endothelium of these vessels.
Collapse
Affiliation(s)
- Samuel N Baldwin
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| | - Shaun L Sandow
- Biomedical Science, School of Health and Sports Science, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Jennifer B Stott
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| |
Collapse
|
14
|
Bein K, Ganguly K, Martin TM, Concel VJ, Brant KA, Di YPP, Upadhyay S, Fabisiak JP, Vuga LJ, Kaminski N, Kostem E, Eskin E, Prows DR, Jang AS, Leikauf GD. Genetic determinants of ammonia-induced acute lung injury in mice. Am J Physiol Lung Cell Mol Physiol 2020; 320:L41-L62. [PMID: 33050709 DOI: 10.1152/ajplung.00276.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In this study, a genetically diverse panel of 43 mouse strains was exposed to ammonia, and genome-wide association mapping was performed employing a single-nucleotide polymorphism (SNP) assembly. Transcriptomic analysis was used to help resolve the genetic determinants of ammonia-induced acute lung injury. The encoded proteins were prioritized based on molecular function, nonsynonymous SNP within a functional domain or SNP within the promoter region that altered expression. This integrative functional approach revealed 14 candidate genes that included Aatf, Avil, Cep162, Hrh4, Lama3, Plcb4, and Ube2cbp, which had significant SNP associations, and Aff1, Bcar3, Cntn4, Kcnq5, Prdm10, Ptcd3, and Snx19, which had suggestive SNP associations. Of these genes, Bcar3, Cep162, Hrh4, Kcnq5, and Lama3 are particularly noteworthy and had pathophysiological roles that could be associated with acute lung injury in several ways.
Collapse
Affiliation(s)
- Kiflai Bein
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Koustav Ganguly
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania.,Unit of Integrated Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Timothy M Martin
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vincent J Concel
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kelly A Brant
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Y P Peter Di
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Swapna Upadhyay
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania.,Unit of Integrated Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - James P Fabisiak
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Louis J Vuga
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Naftali Kaminski
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Medicine, Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Emrah Kostem
- Departments of Computer Science and Human Genetics, University of California, Los Angeles, California
| | - Eleazar Eskin
- Departments of Computer Science and Human Genetics, University of California, Los Angeles, California
| | - Daniel R Prows
- Division of Human Genetics, Cincinnati Children's Hospital and Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Ann-Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - George D Leikauf
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Mondejar-Parreño G, Perez-Vizcaino F, Cogolludo A. Kv7 Channels in Lung Diseases. Front Physiol 2020; 11:634. [PMID: 32676036 PMCID: PMC7333540 DOI: 10.3389/fphys.2020.00634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
Lung diseases constitute a global health concern causing disability. According to WHO in 2016, respiratory diseases accounted for 24% of world population mortality, the second cause of death after cardiovascular diseases. The Kv7 channels family is a group of voltage-dependent K+ channels (Kv) encoded by KCNQ genes that are involved in various physiological functions in numerous cell types, especially, cardiac myocytes, smooth muscle cells, neurons, and epithelial cells. Kv7 channel α-subunits are regulated by KCNE1–5 ancillary β-subunits, which modulate several characteristics of Kv7 channels such as biophysical properties, cell-location, channel trafficking, and pharmacological sensitivity. Kv7 channels are mainly expressed in two large groups of lung tissues: pulmonary arteries (PAs) and bronchial tubes. In PA, Kv7 channels are expressed in pulmonary artery smooth muscle cells (PASMCs); while in the airway (trachea, bronchus, and bronchioles), Kv7 channels are expressed in airway smooth muscle cells (ASMCs), airway epithelial cells (AEPs), and vagal airway C-fibers (VACFs). The functional role of Kv7 channels may vary depending on the cell type. Several studies have demonstrated that the impairment of Kv7 channel has a strong impact on pulmonary physiology contributing to the pathophysiology of different respiratory diseases such as cystic fibrosis, asthma, chronic obstructive pulmonary disease, chronic coughing, lung cancer, and pulmonary hypertension. Kv7 channels are now recognized as playing relevant physiological roles in many tissues, which have encouraged the search for Kv7 channel modulators with potential therapeutic use in many diseases including those affecting the lung. Modulation of Kv7 channels has been proposed to provide beneficial effects in a number of lung conditions. Therefore, Kv7 channel openers/enhancers or drugs acting partly through these channels have been proposed as bronchodilators, expectorants, antitussives, chemotherapeutics and pulmonary vasodilators.
Collapse
Affiliation(s)
- Gema Mondejar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| |
Collapse
|
16
|
Mondéjar-Parreño G, Moral-Sanz J, Barreira B, De la Cruz A, Gonzalez T, Callejo M, Esquivel-Ruiz S, Morales-Cano D, Moreno L, Valenzuela C, Perez-Vizcaino F, Cogolludo A. Activation of K v 7 channels as a novel mechanism for NO/cGMP-induced pulmonary vasodilation. Br J Pharmacol 2019; 176:2131-2145. [PMID: 30883701 DOI: 10.1111/bph.14662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The NO/cGMP pathway represents a major physiological signalling controlling tone in pulmonary arteries (PA), and drugs activating this pathway are used to treat pulmonary arterial hypertension. Kv channels expressed in PA smooth muscle cells (PASMCs) are key determinants of vascular tone. We aimed to analyse the contribution of Kv 1.5 and Kv 7 channels in the electrophysiological and vasodilating effects evoked by NO donors and the GC stimulator riociguat in PA. EXPERIMENTAL APPROACH Kv currents were recorded in isolated rat PASMCs using the patch-clamp technique. Vascular reactivity was assessed in a wire myograph. KEY RESULTS The NO donors diethylamine NONOate diethylammonium (DEA-NO) and sodium nitroprusside hyperpolarized the membrane potential and induced a bimodal effect on Kv currents (augmenting the current between -40 and -10 mV and decreasing it at more depolarized potentials). The hyperpolarization and the enhancement of the current were suppressed by Kv 7 channel inhibitors and by the GC inhibitor ODQ but preserved when Kv 1.5 channels were inhibited. Additionally, DEA-NO enhanced Kv 7.5 currents in COS7 cells expressing the KCNQ5 gene. Riociguat increased Kv currents at all potentials ≥-40 mV and induced membrane hyperpolarization. Both effects were prevented by Kv 7 inhibition. Likewise, PA relaxation induced by NO donors and riociguat was attenuated by Kv 7 inhibitors. CONCLUSIONS AND IMPLICATIONS NO donors and riociguat enhance Kv 7 currents, leading to PASMC hyperpolarization. This mechanism contributes to NO/cGMP-induced PA vasodilation. Our study identifies Kv 7 channels as a novel mechanism of action of vasodilator drugs used in the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Javier Moral-Sanz
- Centres for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Alicia De la Cruz
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Maria Callejo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|