1
|
Chen Y, Xiang S, Chen C, Su Q, Zhang Z, Fan Y, Cui Z, Yin L, Zuo H, Zuo M. Antithrombotic Effect of a Bivalent DNA Aptamer of Thrombin. ACS Biomater Sci Eng 2025; 11:2705-2712. [PMID: 40203196 DOI: 10.1021/acsbiomaterials.5c00222] [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] [Indexed: 04/11/2025]
Abstract
Thrombin plays a critical role in both coagulation and platelet activation, and its interaction with thrombin-protease-activated receptor 1 (PAR1) on platelets and vascular smooth muscle cells (VSMCs) leads to a series of pathological processes such as thrombosis, restenosis, and atherosclerosis. This study investigated the antithrombotic properties of a bivalent DNA aptamer (bApt) with phosphorothioate backbone modification designed to inhibit thrombin, with a specific focus on its ability to regulate the thrombin-PAR1 signaling pathway. The results showed that bApt modulated the thrombin-PAR1 pathway, effectively reduced thrombus formation, platelet aggregation, and VSMC proliferation. Key findings from the study highlight that bApt successfully prolonged coagulation reaction time (R value), coagulation time (K value), maximum amplitude (MA) and reduced coagulation angle (α value), and also prolonged thrombin time (TT) and activated partial thromboplastin time (APTT), in a dose-dependent manner. Moreover, in an arterial injury model, bApt reduced thrombus formation significantly, supporting its potential as a therapeutic agent for thrombotic diseases.
Collapse
MESH Headings
- Aptamers, Nucleotide/pharmacology
- Aptamers, Nucleotide/chemistry
- Thrombin/antagonists & inhibitors
- Thrombin/metabolism
- Animals
- Platelet Aggregation/drug effects
- Fibrinolytic Agents/pharmacology
- Fibrinolytic Agents/chemistry
- Thrombosis/drug therapy
- Humans
- Blood Coagulation/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Male
- Cell Proliferation/drug effects
- Receptor, PAR-1/metabolism
Collapse
Affiliation(s)
- Yanxi Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shoubo Xiang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cardiovascular Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610071, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunfa Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qiuyu Su
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhe Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yangyang Fan
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhihong Cui
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lixue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cardiovascular Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610071, China
| | - Hua Zuo
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Mingliang Zuo
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cardiovascular Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610071, China
| |
Collapse
|
2
|
Li H, Kang L, Dou S, Zhang Y, Zhang Y, Li N, Cao Y, Liu M, Han D, Li K, Feng W. Gleditsiae Sinensis Fructus ingredients and mechanism in anti-asthmatic bronchitis research. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155857. [PMID: 39074420 DOI: 10.1016/j.phymed.2024.155857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Gleditsiae Sinensis Fructus (GSF) is commonly used in traditional medicine to treat respiratory diseases such as bronchial asthma. However, there is a lack of research on the chemical composition of GSF and the pharmacological substance and mechanism of action for GSF in treating bronchial asthma. PURPOSE The chemical constituents of GSF were analyzed using ultrahigh-performance liquid chromatography-quadrupole-Orbitrap high-resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS). In this study, we combined network pharmacology, molecular docking techniques, and experimental validation to explore the therapeutic efficacy and underlying mechanism of GSF in the treatment of bronchial asthma. METHODS Characterization of the chemical constituents of GSF was conducted using UHPLC-Q-Orbitrap HRMS. The identified chemical components were subjected to screening for active ingredients in the Swiss Absorption, Distribution, Metabolism, and Excretion (ADME) database. Relevant databases were utilized to retrieve target proteins for the active ingredients and targets associated with bronchial asthma disease, and the common targets between the two were selected. Subsequently, the protein-protein interaction (PPI) network was constructed using the String database and Cytoscape software to identify key targets. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using the Metascape database. The "component-common target" network was constructed using Cytoscape to identify the primary active ingredients. Molecular docking validation was conducted using AutoDock software. The bronchial asthma mouse model was established using ovalbumin (OVA), and the lung organ index of the mice was measured. Lung tissue pathological changes were observed using hematoxylin and eosin (HE), Periodic Acid-Schiff (PAS), and Masson staining. The respiratory resistance (Penh) of the mice was assessed using a pulmonary function test instrument. An enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of IgE, IL-4, IL-5, and IL-13 in the mouse serum. Immunofluorescence staining was performed to detect the protein expression levels of AKT and PI3K in the lung tissues. An in vitro experiment was performed to observe the effects of echinocystic acid (EA) on IL-4 stimulated Human ASMCs (hASMCs). Cell viability was measured using a CCK-8 assay to calculate the IC50 value of the EA. A wound healing test was conducted to observe the effect of EA on degree of healing. RT-qPCR was performed to detect the influence of EA on the mRNA expression levels of ALB, SRC, TNF-α, AKT1, and IL6 in the cells. RESULTS A total of 95 chemical constituents were identified from the GSF. Of these, 37 were identified as active ingredients. There were 169 overlapping targets between the active ingredients and the disease targets. A topological analysis of the protein-protein interaction (PPI) network identified the core targets as IL6, TNF, ALB, AKT1, and SRC. An enrichment analysis revealed that the treatment of bronchial asthma with GSF primarily involved the AGE-RAGE signaling pathway and the PI3K-Akt signaling pathway, among others. The primary active ingredients included 13(s)-HOTRE, linolenic acid, and acacetin. The molecular docking results demonstrated a favorable binding activity between the critical components of GSF and the core targets. Animal experimental studies indicated that GSF effectively improved symptoms, lung function, and lung tissue pathological changes in the OVA-induced asthmatic mice, while alleviating inflammatory responses. GSF decreased the fluorescent intensity of the AKT and PI3K proteins. The IC50 value of EA was 30.02μg/ml. EA (30) significantly promoted the proliferation of IL4-stimulated hASMCs cells. EA (30) significantly increased the expression of ALB and SRC mRNA and decreased the expressions of TNF-α, AKT, and IL6 mRNA. CONCLUSION The multiple active ingredients found in GSF exerted their anti-inflammatory effects through multiple targets and pathways. This preliminary study revealed the core target and the mechanism of action underlying its treatment of bronchial asthma. These findings provided valuable insights for further research on the pharmacological substances and quality control of GSF.
Collapse
Affiliation(s)
- Hongwei Li
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Le Kang
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou 450046, China.
| | - Shirong Dou
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Yiming Zhang
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Yumei Zhang
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Ning Li
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Yangang Cao
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Mengyun Liu
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Deen Han
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Kai Li
- Henan University of Chinese Medicine, Zhengzhou 450046, China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China.
| | - Weisheng Feng
- Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
3
|
Sung DJ, Park S, Noh HJ, Golpasandi S, Eun SH, Lee H, Kim B, Wie J, Seo MS, Park SW, Bae YM. Receptor-specific contributions of caveolae, PKC, and Src tyrosine kinase to serotonergic and adrenergic regulation of Kv channels and vasoconstriction. Life Sci 2023; 328:121903. [PMID: 37394095 DOI: 10.1016/j.lfs.2023.121903] [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: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
AIMS Caveolae are invaginated, Ω-shaped membrane structures. They are now recognized as portals for signal transduction of multiple chemical and mechanical stimuli. Notably, the contribution of caveolae has been reported to be receptor-specific. However, details of how they differentially contribute to receptor signaling remain unclear. MAIN METHODS Using isometric tension measurements, patch-clamping, and western blotting, we examined the contribution of caveolae and their related signaling pathways to serotonergic (5-HT2A receptor-mediated) and adrenergic (α1-adrenoceptor-mediated) signaling in rat mesenteric arteries. KEY FINDINGS Disruption of caveolae by methyl-β-cyclodextrin effectively blocked vasoconstriction mediated by the 5-HT2A receptor (5-HT2AR), but not by the α1-adrenoceptor. Caveolar disruption selectively impaired 5-HT2AR-mediated voltage-dependent K+ channel (Kv) inhibition, but not α1-adrenoceptor-mediated Kv inhibition. In contrast, both serotonergic and α1-adrenergic effects on vasoconstriction, as well as Kv currents, were similarly blocked by the Src tyrosine kinase inhibitor PP2. However, inhibition of protein kinase C (PKC) by either GO6976 or chelerythrine selectively attenuated the effects mediated by the α1-adrenoceptor, but not by 5-HT2AR. Disruption of caveolae decreased 5-HT2AR-mediated Src phosphorylation, but not α1-adrenoceptor-mediated Src phosphorylation. Finally, the PKC inhibitor GO6976 blocked Src phosphorylation by the α1-adrenoceptor, but not by 5-HT2AR. SIGNIFICANCE 5-HT2AR-mediated Kv inhibition and vasoconstriction are dependent on caveolar integrity and Src tyrosine kinase, but not on PKC. In contrast, α1-adrenoceptor-mediated Kv inhibition and vasoconstriction are not dependent on caveolar integrity, but rather on PKC and Src tyrosine kinase. Caveolae-independent PKC is upstream of Src activation for α1-adrenoceptor-mediated Kv inhibition and vasoconstriction.
Collapse
Affiliation(s)
- Dong Jun Sung
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; Sports Convergence Institute, Konkuk University, Chungju 27478, Republic of Korea; Center for Metabolic Diseases, Konkuk University, Chungju 27478, Republic of Korea; Research Institute for Biomedical & Health Science, Chungju 27478, Republic of Korea
| | - Solah Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyun Ju Noh
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Shadi Golpasandi
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Seo Hyeon Eun
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyeryeong Lee
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Jinhong Wie
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Mi Seon Seo
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, Seongnam 13135, Republic of Korea.
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea.
| |
Collapse
|
4
|
Zhang YL, Zhang RG, Chen FY, Qiu ZE, Chen L, Huang ZX, Huang J, Zhu YX, Zhao L, Zhou WL. Cellular Mechanism Underlying the Facilitation of Contractile Response Induced by Tumor Necrosis Factor-α in Mouse Tracheal Smooth Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 192:104-111. [PMID: 34756873 DOI: 10.1016/j.ajpath.2021.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 01/10/2023]
Abstract
The proinflammatory cytokine tumor necrosis factor-α (TNF-α) augments intracellular Ca2+ signaling and contractile responses of airway smooth muscles, leading to airway hyperresponsiveness. However, the underlying mechanism has not been fully elucidated. This study aimed to investigate the cellular mechanism of the potentiated contraction of mouse tracheal smooth muscle induced by TNF-α. The results showed that TNF-α triggered facilitation of mouse tracheal smooth muscle contraction in an epithelium-independent manner. The TNF-α-induced hypercontractility could be suppressed by the protein kinase C inhibitor GF109203X, the tyrosine kinase inhibitor genistein, the Src inhibitor PP2, or the L-type voltage-dependent Ca2+ channel blocker nifedipine. After TNF-α incubation, the α1C L-type Ca2+ channel (CaV1.2) was up-regulated in primary cultured mouse tracheal smooth muscle cells. Pronounced phosphotyrosine levels also were observed in mouse tracheas. In conclusion, this study showed that TNF-α enhanced airway smooth muscle contraction via protein kinase C-Src-CaV1.2 pathways, which provides novel insights into the pathologic role of proinflammatory cytokines in mediating airway hyperresponsiveness.
Collapse
Affiliation(s)
- Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui-Gang Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Basic Medical School, Guangdong Medical University, Zhanjiang, China
| | - Feng-Ying Chen
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pathology, The Maternal and Child Health Care Hospital of HuaDu District (Huzhong Hospital), Guangzhou, China
| | - Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jiehong Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhao
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Tian WM, Chen WW, Chen Y, Lin CX, Huang JX, Song YP, Yang YG. The role and mechanism of 1,25-dihydroxyvitamin D 3 in regulating the Rho-kinase signaling pathway in asthmatic rats. Transl Pediatr 2021; 10:773-782. [PMID: 34012827 PMCID: PMC8107869 DOI: 10.21037/tp-20-365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Bronchial asthma (referred to as asthma in the present study) is the most common chronic airway inflammatory disease in childhood. The present study aimed to investigate the effect of 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] on VDR expression, which is closely associated with asthmatic airway smooth muscle cells (ASMCs), and explored its role and mechanism in the Rho-kinase signaling pathway. METHODS The acute asthma model was induced by ovalbumin (OVA) and pertussis bacillus, and ASMCs obtained from asthmatic rats were cultured in vitro. These cells were randomly divided into five groups: control (N) group, TNF-α (TNF) group, 1,25-(OH)2D3 (VD) group, dexamethasone (DXM) group, and 1,25-(OH)2D3 + DXM (L) group. The protein expression levels of VDR, ROCK, MLC20 and P-MLC20 were detected by western blot, and the mRNA expression levels of VDR, ROCK, MLC20 and P-MLC20 were detected by real-time quantitative PCR. RESULTS The expression of ROCK, MLC20 and P-MLC20 in each treatment group were significantly lower, when compared to the TNF group (P<0.05), but this remained stronger than (P<0.05) or similar to (P>0.05) that in the N group. CONCLUSIONS The regulation mechanism of 1,25-(OH)2D3 in alleviating asthma should be correlated to its regulation of the expression of related signaling molecules in the Rho-kinase signaling pathway, and this effect may be achieved by regulating the mRNA and protein expression of the VDR gene.
Collapse
Affiliation(s)
- Wei-Min Tian
- Department of Pediatrics, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei-Wei Chen
- Department of Pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yu Chen
- Department of Pediatrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Chen-Xi Lin
- Department of Pediatrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Jin-Xian Huang
- Department of Pediatrics, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ya-Ping Song
- School of Medicine, Xiamen University, Xiamen, China
| | - Yun-Gang Yang
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
6
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
7
|
Yoo EJ, Cao G, Koziol‐White CJ, Ojiaku CA, Sunder K, Jude JA, Michael JV, Lam H, Pushkarsky I, Damoiseaux R, Di Carlo D, Ahn K, An SS, Penn RB, Panettieri RA. Gα 12 facilitates shortening in human airway smooth muscle by modulating phosphoinositide 3-kinase-mediated activation in a RhoA-dependent manner. Br J Pharmacol 2017; 174:4383-4395. [PMID: 28921504 PMCID: PMC5715591 DOI: 10.1111/bph.14040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE PI3K-dependent activation of Rho kinase (ROCK) is necessary for agonist-induced human airway smooth muscle cell (HASMC) contraction, and inhibition of PI3K promotes bronchodilation of human small airways. The mechanisms driving agonist-mediated PI3K/ROCK axis activation, however, remain unclear. Given that G12 family proteins activate ROCK pathways in other cell types, their role in M3 muscarinic acetylcholine receptor-stimulated PI3K/ROCK activation and contraction was examined. EXPERIMENTAL APPROACH Gα12 coupling was evaluated using co-immunoprecipitation and serum response element (SRE)-luciferase reporter assays. siRNA and pharmacological approaches, as well as overexpression of a regulator of G-protein signaling (RGS) proteins were applied in HASMCs. Phosphorylation levels of Akt, myosin phosphatase targeting subunit-1 (MYPT1), and myosin light chain-20 (MLC) were measured. Contraction and shortening were evaluated using magnetic twisting cytometry (MTC) and micro-pattern deformation, respectively. Human precision-cut lung slices (hPCLS) were utilized to evaluate bronchoconstriction. KEY RESULTS Knockdown of M3 receptors or Gα12 attenuated activation of Akt, MYPT1, and MLC phosphorylation. Gα12 coimmunoprecipitated with M3 receptors, and p115RhoGEF-RGS overexpression inhibited carbachol-mediated induction of SRE-luciferase reporter. p115RhoGEF-RGS overexpression inhibited carbachol-induced activation of Akt, HASMC contraction, and shortening. Moreover, inhibition of RhoA blunted activation of PI3K. Lastly, RhoA inhibitors induced dilation of hPCLS. CONCLUSIONS AND IMPLICATIONS Gα12 plays a crucial role in HASMC contraction via RhoA-dependent activation of the PI3K/ROCK axis. Inhibition of RhoA activation induces bronchodilation in hPCLS, and targeting Gα12 signaling may elucidate novel therapeutic targets in asthma. These findings provide alternative approaches to the clinical management of airway obstruction in asthma.
Collapse
Affiliation(s)
- Edwin J Yoo
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - Cynthia J Koziol‐White
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - Christie A Ojiaku
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - Krishna Sunder
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - Joseph A Jude
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| | - James V Michael
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung CenterThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Hong Lam
- Department of Environmental Health and EngineeringJohns Hopkins Bloomberg School of Public HealthBaltimoreMDUSA
| | - Ivan Pushkarsky
- Department of BioengineeringUniversity of CaliforniaLos AngelesCAUSA
| | - Robert Damoiseaux
- Department of Molecular and Medicinal PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCAUSA
| | - Dino Di Carlo
- Department of BioengineeringUniversity of CaliforniaLos AngelesCAUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCAUSA
- Department of Mechanical EngineeringUniversity of CaliforniaLos AngelesCAUSA
| | - Kwangmi Ahn
- National Institute of Mental HealthBethesdaMDUSA
| | - Steven S An
- Department of Environmental Health and EngineeringJohns Hopkins Bloomberg School of Public HealthBaltimoreMDUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMDUSA
| | - Raymond B Penn
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung CenterThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health InstituteRutgers UniversityNew BrunswickNJUSA
| |
Collapse
|
8
|
Shaifta Y, MacKay CE, Irechukwu N, O'Brien KA, Wright DB, Ward JPT, Knock GA. Transforming growth factor-β enhances Rho-kinase activity and contraction in airway smooth muscle via the nucleotide exchange factor ARHGEF1. J Physiol 2017; 596:47-66. [PMID: 29071730 PMCID: PMC5746525 DOI: 10.1113/jp275033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/19/2017] [Indexed: 01/16/2023] Open
Abstract
Key points Transforming growth‐factor‐β (TGF‐β) and RhoA/Rho‐kinase are independently implicated in the airway hyper‐responsiveness associated with asthma, but how these proteins interact is not fully understood. We examined the effects of pre‐treatment with TGF‐β on expression and activity of RhoA, Rho‐kinase and ARHGEF1, an activator of RhoA, as well as on bradykinin‐induced contraction, in airway smooth muscle. TGF‐β enhanced bradykinin‐induced RhoA translocation, Rho‐kinase‐dependent phosphorylation and contraction, but partially suppressed bradykinin‐induced RhoA activity (RhoA‐GTP content). TGF‐β enhanced the expression of ARHGEF1, while a small interfering RNA against ARHGEF1 and a RhoGEF inhibitor prevented the effects of TGF‐β on RhoA and Rho‐kinase activity and contraction, respectively. ARHGEF1 expression was also enhanced in airway smooth muscle from asthmatic patients and ovalbumin‐sensitized mice. ARHGEF1 is a key TGF‐β target gene, an important regulator of Rho‐kinase activity and therefore a potential therapeutic target for the treatment of asthmatic airway hyper‐responsiveness.
Abstract Transforming growth factor‐β (TGF‐β), RhoA/Rho‐kinase and Src‐family kinases (SrcFK) have independently been implicated in airway hyper‐responsiveness, but how they interact to regulate airway smooth muscle contractility is not fully understood. We found that TGF‐β pre‐treatment enhanced acute contractile responses to bradykinin (BK) in isolated rat bronchioles, and inhibitors of RhoGEFs (Y16) and Rho‐kinase (Y27632), but not the SrcFK inhibitor PP2, prevented this enhancement. In cultured human airway smooth muscle cells (hASMCs), TGF‐β pre‐treatment enhanced the protein expression of the Rho guanine nucleotide exchange factor ARHGEF1, MLC20, MYPT‐1 and the actin‐severing protein cofilin, but not of RhoA, ROCK2 or c‐Src. In hASMCs, acute treatment with BK triggered subcellular translocation of ARHGEF1 and RhoA and enhanced auto‐phosphorylation of SrcFK and phosphorylation of MYPT1 and MLC20, but induced de‐phosphorylation of cofilin. TGF‐β pre‐treatment amplified the effects of BK on RhoA translocation and MYPT1/MLC20 phosphorylation, but suppressed the effects of BK on RhoA‐GTP content, SrcFK auto‐phosphorylation and cofilin de‐phosphorylation. In hASMCs, an ARHGEF1 small interfering RNA suppressed the effects of BK and TGF‐β on RhoA‐GTP content, RhoA translocation and MYPT1 and MLC20 phosphorylation, but minimally influenced the effects of TGF‐β on cofilin expression and phosphorylation. ARHGEF1 expression was also enhanced in ASMCs of asthmatic patients and in lungs of ovalbumin‐sensitized mice. Our data indicate that TGF‐β enhances BK‐induced contraction, RhoA translocation and Rho‐kinase activity in airway smooth muscle largely via ARHGEF1, but independently of SrcFK and total RhoA‐GTP content. A role for smooth muscle ARHGEF1 in asthmatic airway hyper‐responsiveness is worthy of further investigation. Transforming growth‐factor‐β (TGF‐β) and RhoA/Rho‐kinase are independently implicated in the airway hyper‐responsiveness associated with asthma, but how these proteins interact is not fully understood. We examined the effects of pre‐treatment with TGF‐β on expression and activity of RhoA, Rho‐kinase and ARHGEF1, an activator of RhoA, as well as on bradykinin‐induced contraction, in airway smooth muscle. TGF‐β enhanced bradykinin‐induced RhoA translocation, Rho‐kinase‐dependent phosphorylation and contraction, but partially suppressed bradykinin‐induced RhoA activity (RhoA‐GTP content). TGF‐β enhanced the expression of ARHGEF1, while a small interfering RNA against ARHGEF1 and a RhoGEF inhibitor prevented the effects of TGF‐β on RhoA and Rho‐kinase activity and contraction, respectively. ARHGEF1 expression was also enhanced in airway smooth muscle from asthmatic patients and ovalbumin‐sensitized mice. ARHGEF1 is a key TGF‐β target gene, an important regulator of Rho‐kinase activity and therefore a potential therapeutic target for the treatment of asthmatic airway hyper‐responsiveness.
Collapse
Affiliation(s)
- Yasin Shaifta
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Charles E MacKay
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Nneka Irechukwu
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Katie A O'Brien
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - David B Wright
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Greg A Knock
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| |
Collapse
|
9
|
MacKay CE, Shaifta Y, Snetkov VV, Francois AA, Ward JPT, Knock GA. ROS-dependent activation of RhoA/Rho-kinase in pulmonary artery: Role of Src-family kinases and ARHGEF1. Free Radic Biol Med 2017; 110:316-331. [PMID: 28673614 PMCID: PMC5542024 DOI: 10.1016/j.freeradbiomed.2017.06.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022]
Abstract
The role of reactive oxygen species (ROS) in smooth muscle contraction is poorly understood. We hypothesised that G-protein coupled receptor (GPCR) activation and hypoxia induce Rho-kinase activity and contraction in rat intra-pulmonary artery (IPA) via stimulation of ROS production and subsequent Src-family kinase (SrcFK) activation. The T-type prostanoid receptor agonist U46619 induced ROS production in pulmonary artery smooth muscle cells (PASMC). U46619 also induced c-Src cysteine oxidation, SrcFK auto-phosphorylation, MYPT-1 and MLC20 phosphorylation and contraction in IPA, and all these responses were inhibited by antioxidants (ebselen, Tempol). Contraction and SrcFK/MYPT-1/MLC20 phosphorylations were also inhibited by combined superoxide dismutase and catalase, or by the SrcFK antagonist PP2, while contraction and MYPT-1/MLC20 phosphorylations were inhibited by the Rho guanine nucleotide exchange factor (RhoGEF) inhibitor Y16. H2O2 and the superoxide-generating quinoledione LY83583 both induced c-Src oxidation, SrcFK auto-phosphorylation and contraction in IPA. LY83583 and H2O2-induced contractions were inhibited by PP2, while LY83583-induced contraction was also inhibited by antioxidants and Y16. SrcFK auto-phosphorylation and MYPT-1/MLC20 phosphorylation was also induced by hypoxia in IPA and this was blocked by mitochondrial inhibitors rotenone and myxothiazol. In live PASMC, sub-cellular translocation of RhoA and the RhoGEF ARHGEF1 was triggered by both U46619 and LY83583 and this translocation was blocked by antioxidants and PP2. RhoA translocation was also inhibited by an ARHGEF1 siRNA. U46619 enhanced ROS-dependent co-immunoprecipitation of ARHGEF1 with c-Src. Our results demonstrate a link between GPCR-induced cytosolic ROS or hypoxia-induced mitochondrial ROS and SrcFK activity, Rho-kinase activity and contraction. ROS and SrcFK activate RhoA via ARHGEF1.
Collapse
Affiliation(s)
- Charles E MacKay
- Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Yasin Shaifta
- Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Vladimir V Snetkov
- Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Asvi A Francois
- Cardiovascular Division, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Jeremy P T Ward
- Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Greg A Knock
- Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.
| |
Collapse
|
10
|
Ferland DJ, Darios ES, Neubig RR, Sjögren B, Truong N, Torres R, Dexheimer TS, Thompson JM, Watts SW. Chemerin-induced arterial contraction is G i- and calcium-dependent. Vascul Pharmacol 2016; 88:30-41. [PMID: 27890480 DOI: 10.1016/j.vph.2016.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/27/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022]
Abstract
Chemerin is an adipokine associated with increased blood pressure, and may link obesity with hypertension. We tested the hypothesis that chemerin-induced contraction of the vasculature occurs via calcium flux in smooth muscle cells. Isometric contraction of rat aortic rings was performed in parallel with calcium kinetics of rat aortic smooth muscle cells to assess the possible signaling pathway. Chemerin-9 (nonapeptide of the chemerin S157 isoform) caused a concentration-dependent contraction of isolated aorta (EC50 100nM) and elicited a concentration-dependent intracellular calcium response (EC50 10nM). Pertussis toxin (Gi inhibitor), verapamil (L-type Ca2+ channel inhibitor), PP1 (Src inhibitor), and Y27632 (Rho kinase inhibitor) reduced both calcium influx and isometric contraction to chemerin-9 but PD098059 (Erk MAPK inhibitor) and U73122 (PLC inhibitor) had little to no effect on either measure of chemerin signaling. Although our primary aim was to examine chemerin signaling, we also highlight differences in the mechanisms of chemerin-9 and recombinant chemerin S157. These data support a chemerin-induced contractile mechanism in vascular smooth muscle that functions through Gi proteins to activate L-type Ca2+ channels, Src, and Rho kinase. There is mounting evidence linking chemerin to hypertension and this mechanism brings us closer to targeting chemerin as a form of therapy.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Calcium/metabolism
- Calcium Channels, L-Type/metabolism
- Chemokines/administration & dosage
- Chemokines/metabolism
- Dose-Response Relationship, Drug
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Hypertension/physiopathology
- Intercellular Signaling Peptides and Proteins/administration & dosage
- Intercellular Signaling Peptides and Proteins/metabolism
- Male
- Muscle Contraction/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- David J Ferland
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States.
| | - Emma S Darios
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Benita Sjögren
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Nguyen Truong
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Rosa Torres
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Thomas S Dexheimer
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Janice M Thompson
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, 1355 Bogue Street Rm B445, Michigan State University, East Lansing, MI 48824-1317, United States
| |
Collapse
|
11
|
Barnes PJ. Kinases as Novel Therapeutic Targets in Asthma and Chronic Obstructive Pulmonary Disease. Pharmacol Rev 2016; 68:788-815. [PMID: 27363440 DOI: 10.1124/pr.116.012518] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multiple kinases play a critical role in orchestrating the chronic inflammation and structural changes in the respiratory tract of patients with asthma and chronic obstructive pulmonary disease (COPD). Kinases activate signaling pathways that lead to contraction of airway smooth muscle and release of inflammatory mediators (such as cytokines, chemokines, growth factors) as well as cell migration, activation, and proliferation. For this reason there has been great interest in the development of kinase inhibitors as anti-inflammatory therapies, particular where corticosteroids are less effective, as in severe asthma and COPD. However, it has proven difficult to develop selective kinase inhibitors that are both effective and safe after oral administration and this has led to a search for inhaled kinase inhibitors, which would reduce systemic exposure. Although many kinases have been implicated in inflammation and remodeling of airway disease, very few classes of drug have reached the stage of clinical studies in these diseases. The most promising drugs are p38 MAP kinases, isoenzyme-selective PI3-kinases, Janus-activated kinases, and Syk-kinases, and inhaled formulations of these drugs are now in development. There has also been interest in developing inhibitors that block more than one kinase, because these drugs may be more effective and with less risk of losing efficacy with time. No kinase inhibitors are yet on the market for the treatment of airway diseases, but as kinase inhibitors are improved from other therapeutic areas there is hope that these drugs may eventually prove useful in treating refractory asthma and COPD.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
12
|
Shaifta Y, Irechukwu N, Prieto-Lloret J, MacKay CE, Marchon KA, Ward JPT, Knock GA. Divergent modulation of Rho-kinase and Ca(2+) influx pathways by Src family kinases and focal adhesion kinase in airway smooth muscle. Br J Pharmacol 2015; 172:5265-80. [PMID: 26294392 PMCID: PMC4864488 DOI: 10.1111/bph.13313] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/02/2015] [Accepted: 08/19/2015] [Indexed: 02/06/2023] Open
Abstract
Background and Purpose The importance of tyrosine kinases in airway smooth muscle (ASM) contraction is not fully understood. The aim of this study was to investigate the role of Src‐family kinases (SrcFK) and focal adhesion kinase (FAK) in GPCR‐mediated ASM contraction and associated signalling events. Experimental Approach Contraction was recorded in intact or α‐toxin permeabilized rat bronchioles. Phosphorylation of SrcFK, FAK, myosin light‐chain‐20 (MLC20) and myosin phosphatase targeting subunit‐1 (MYPT‐1) was evaluated in cultured human ASM cells (hASMC). [Ca2+]i was evaluated in Fura‐2 loaded hASMC. Responses to carbachol (CCh) and bradykinin (BK) and the contribution of SrcFK and FAK to these responses were determined. Key Results Contractile responses in intact bronchioles were inhibited by antagonists of SrcFK, FAK and Rho‐kinase, while after α‐toxin permeabilization, they were sensitive to inhibition of SrcFK and Rho‐kinase, but not FAK. CCh and BK increased phosphorylation of MYPT‐1 and MLC20 and auto‐phosphorylation of SrcFK and FAK. MYPT‐1 phosphorylation was sensitive to inhibition of Rho‐kinase and SrcFK, but not FAK. Contraction induced by SR Ca2+ depletion and equivalent [Ca2+]i responses in hASMC were sensitive to inhibition of both SrcFK and FAK, while depolarization‐induced contraction was sensitive to FAK inhibition only. SrcFK auto‐phosphorylation was partially FAK‐dependent, while FAK auto‐phosphorylation was SrcFK‐independent. Conclusions and Implications SrcFK mediates Ca2+‐sensitization in ASM, while SrcFK and FAK together and individually influence multiple Ca2+ influx pathways. Tyrosine phosphorylation is therefore a key upstream signalling event in ASM contraction and may be a viable target for modulating ASM tone in respiratory disease.
Collapse
Affiliation(s)
- Yasin Shaifta
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Nneka Irechukwu
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jesus Prieto-Lloret
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Charles E MacKay
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Keisha A Marchon
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Greg A Knock
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|