1
|
Dong L, Klassen TP, Johnson DW, Correll R, Gouin S, Bhatt M, Patel H, Joubert G, Black KJL, Turner TWS, Whitehouse SR, Plint AC, Heath A. The probability of reducing hospitalization rates for bronchiolitis with epinephrine and dexamethasone: A Bayesian analysis. PLoS One 2025; 20:e0318853. [PMID: 40378135 PMCID: PMC12083810 DOI: 10.1371/journal.pone.0318853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 04/16/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Bronchiolitis exerts a high burden on children, their families and the healthcare system. The Canadian Bronchiolitis Epinephrine Steroid Trial (CanBEST) assessed whether administering epinephrine alone, dexamethasone alone, or in combination (EpiDex) could reduce bronchiolitis-related hospitalizations among children less than 12 months of age compared to placebo. CanBEST demonstrated a statistically significant reduction in 7-day hospitalization risk with EpiDex in an unadjusted analysis but not after adjustment. OBJECTIVE To explore the probability that EpiDex results in a reduction in hospitalizations using Bayesian methods. STUDY DESIGN Using prior distributions that represent varying levels of preexisting enthusiasm or skepticism, i.e., how confident or doubtful one is that EpiDex may reduce hospitalizations, and information about the treatment effect before data were collected, the posterior distribution of the relative risk of hospitalization compared to placebo was determined. The probability that the treatment effect is less than 1, 0.9, 0.8 and 0.6, indicating increasing reductions in hospitalization risk, are computed alongside 95% credible intervals. RESULTS Combining a minimally informative prior distribution with the data from CanBEST provides comparable results to the original analysis. Unless strongly skeptical views about the effectiveness of EpiDex were considered, the 95% credible interval for the treatment effect lies below 1, indicating a reduction in hospitalizations. There is a 90% probability that EpiDex results in a clinically meaningful reduction in hospitalization of 10% even when incorporating skeptical views, with a 67% probability when considering strongly skeptical views. CONCLUSION A Bayesian analysis demonstrates a high chance that EpiDex reduces hospitalization rates for bronchiolitis, although strongly skeptical individuals may require additional evidence to change practice. TRIAL REGISTRATION Clinical Trial registry name, registration number: Current Controlled Trials number, ISRCTN56745572.
Collapse
Affiliation(s)
- Larry Dong
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Canada
| | - Terry P. Klassen
- Children’s Hospital Research Institute of Manitoba, Department of Pediatrics and Child Health, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David W. Johnson
- Alberta Children’s Hospital Research Institute, Calgary, Canada
- Departments of Pediatrics, Emergency Medicine and Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Rhonda Correll
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Serge Gouin
- Pediatric Emergency Department, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Canada
- Department of Pediatrics, Université de Montréal, Montréal, Canada
| | - Maala Bhatt
- Department of Pediatrics, University of Ottawa, Ottawa, Canada
- Children’s Hospital of Eastern Ontario, Ottawa, Canada
| | - Hema Patel
- McGill University Health Centre Research Institute, Montréal, Canada
- Department of Pediatrics, McGill University, Montreal, Canada
| | - Gary Joubert
- Department of Pediatrics, Children’s Hospital, Western University, London, Canada
| | - Karen J. L. Black
- Division of Pediatric Emergency Medicine, British Columbia Children’s Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Troy W. S. Turner
- Pediatric Emergency Department, Stollery Children’s HospitalAlberta, Canada
- Department of Pediatrics, University of Alberta. Edmonton, Alberta, Canada
| | | | - Amy C. Plint
- Children’s Hospital of Eastern Ontario, Ottawa, Canada
- Department of Pediatrics and Emergency Medicine, University of Ottawa, Ottawa, Canada
| | - Anna Heath
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Canada
- Department of Statistical Science, University College London, London, United Kingdom
| |
Collapse
|
2
|
Joshi JC, Joshi B, Zhang C, Banerjee S, Vellingiri V, Raghunathrao VAB, Anwar M, Rokade TP, Zhang L, Amin R, Song Y, Mehta D. RGS2 is an innate immune checkpoint for suppressing Gαq-mediated IFNγ generation and lung injury. iScience 2025; 28:111878. [PMID: 40041768 PMCID: PMC11876898 DOI: 10.1016/j.isci.2025.111878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/01/2024] [Accepted: 01/20/2025] [Indexed: 03/12/2025] Open
Abstract
Interferon gamma (IFNγ), a type II interferon, augments tissue inflammation following infections, leading to lethal acute lung injury (ALI), yet the mechanisms controlling IFNγ generation in the lungs remain elusive. Here, we identified regulator of G protein signaling 2 (RGS2) as a gatekeeper of the lung's IFNγ levels during infections. Deletion of RGS2 sustained an increase in IFNγ levels in macrophages, leading to unresolvable inflammatory lung injury. This response was not seen in RGS2 null chimeric mice receiving wild-type (WT) bone marrow or the RGS2 gene in alveolar macrophages (AMs) or IFNγ-blocking antibody. RGS2 functioned by suppressing Gαq-mediated IFNγ generation and AM inflammatory signaling. Thus, the inhibition of Gαq blocked IFNγ generation in AMs and rewired AM transcriptomes from an inflammatory to a reparative phenotype in RGS2 null mice, pointing to the RGS2-Gαq axis as a potential target for suppressing inflammatory injury.
Collapse
Affiliation(s)
- Jagdish Chandra Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
- Lake Erie College of Osteopathic Medicine, School of Pharmacy, Erie, PA, USA
| | - Bhagwati Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Somenath Banerjee
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vigneshwaran Vellingiri
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vijay Avin Balaji Raghunathrao
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Mumtaz Anwar
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Tejas Pravin Rokade
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Vascular Medicine Institute, Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ruhul Amin
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dolly Mehta
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Calzetta L, Page C, Matera MG, Cazzola M, Rogliani P. Drug-Drug Interactions and Synergy: From Pharmacological Models to Clinical Application. Pharmacol Rev 2024; 76:1159-1220. [PMID: 39009470 DOI: 10.1124/pharmrev.124.000951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
This review explores the concept of synergy in pharmacology, emphasizing its importance in optimizing treatment outcomes through the combination of drugs with different mechanisms of action. Synergy, defined as an effect greater than the expected additive effect elicited by individual agents according to specific predictive models, offers a promising approach to enhance therapeutic efficacy while minimizing adverse events. The historical evolution of synergy research, from ancient civilizations to modern pharmacology, highlights the ongoing quest to understand and harness synergistic interactions. Key concepts, such as concentration-response curves, additive effects, and predictive models, are discussed in detail, emphasizing the need for accurate assessment methods throughout translational drug development. Although various mathematical models exist for synergy analysis, selecting the appropriate model and software tools remains a challenge, necessitating careful consideration of experimental design and data interpretation. Furthermore, this review addresses practical considerations in synergy assessment, including preclinical and clinical approaches, mechanism of action, and statistical analysis. Optimizing synergy requires attention to concentration/dose ratios, target site localization, and timing of drug administration, ensuring that the benefits of combination therapy detected bench-side are translatable into clinical practice. Overall, the review advocates for a systematic approach to synergy assessment, incorporating robust statistical analysis, effective and simplified predictive models, and collaborative efforts across pivotal sectors, such as academic institutions, pharmaceutical companies, and regulatory agencies. By overcoming critical challenges and maximizing therapeutic potential, effective synergy assessment in drug development holds promise for advancing patient care. SIGNIFICANCE STATEMENT: Combining drugs with different mechanisms of action for synergistic interactions optimizes treatment efficacy and safety. Accurate interpretation of synergy requires the identification of the expected additive effect. Despite innovative models to predict the additive effect, consensus in drug-drug interactions research is lacking, hindering the bench-to-bedside development of combination therapies. Collaboration among science, industry, and regulation is crucial for advancing combination therapy development, ensuring rigorous application of predictive models in clinical settings.
Collapse
Affiliation(s)
- Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Clive Page
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Maria Gabriella Matera
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Mario Cazzola
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| | - Paola Rogliani
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (L.C.); Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, United Kingdom (C.P.); Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.G.-M.); and Respiratory Medicine Unit, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy (M.C., P.R.)
| |
Collapse
|
4
|
Mohammed SAH, Mirdamadi M, Szucs KF, Gaspar R. Non-genomic actions of steroid hormones on the contractility of non-vascular smooth muscles. Biochem Pharmacol 2024; 222:116063. [PMID: 38373593 DOI: 10.1016/j.bcp.2024.116063] [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: 10/01/2023] [Revised: 01/03/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Steroid hormones play an important role in physiological processes. The classical pathway of steroid actions is mediated by nuclear receptors, which regulate genes to modify biological processes. Non-genomic pathways of steroid actions are also known, mediated by cell membrane-located seven transmembrane domain receptors. Sex steroids and glucocorticoids have several membrane receptors already identified to mediate their rapid actions. However, mineralocorticoids have no identified membrane receptors, although their rapid actions are also measurable. In non-vascular smooth muscles (bronchial, uterine, gastrointestinal, and urinary), the rapid actions of steroids are mediated through the modification of the intracellular Ca2+ level by various Ca-channels and the cAMP and IP3 system. The non-genomic action can be converted into a genomic one, suggesting that these distinct pathways may interconnect, resulting in convergence between them. Sex steroids mostly relax all the non-vascular smooth muscles, except androgens and progesterone, which contract colonic and urinary bladder smooth muscles, respectively. Corticosteroids also induce relaxation in bronchial and uterine tissues, but their actions on gastrointestinal and urinary bladder smooth muscles have not been investigated yet. Bile acids also contribute to the smooth muscle contractility. Although the therapeutic application of the rapid effects of steroid hormones and their analogues for smooth muscle contractility disorders seems remote, the actions and mechanism discovered so far are promising. Further research is needed to expand our knowledge in this field by using existing experience. One of the greatest challenges is to separate genomic and non-genomic effects, but model molecules are available to start this line of research.
Collapse
Affiliation(s)
- Saif-Alnasr H Mohammed
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Mohsen Mirdamadi
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Kalman F Szucs
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Robert Gaspar
- Department of Pharmacology and Pharmacotherapy, Albert-Szent-Györgyi Medical School, University of Szeged, Hungary.
| |
Collapse
|
5
|
Seaton WB, Burke SJ, Fisch AR, Schilletter WA, Beck MGA, Cassagne GA, Harvey I, Fontenot MS, Collier JJ, Campagna SR. Channel Expansion in the Ligand-Binding Domain of the Glucocorticoid Receptor Contributes to the Activity of Highly Potent Glucocorticoid Analogues. Molecules 2024; 29:1546. [PMID: 38611825 PMCID: PMC11013598 DOI: 10.3390/molecules29071546] [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: 03/15/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Glucocorticoids (GCs) act through the glucocorticoid receptor (GR) and are commonly used as anti-inflammatory and immunosuppressant medications. Chronic GC use has been linked with unwanted complications such as steroid-induced diabetes mellitus (SIDM), although the mechanisms for these effects are not completely understood. Modification of six GC parent molecules with 2-mercaptobenzothiazole resulted in consistently less promoter activity in transcriptional activation assays using a 3xGRE reporter construct while constantly reducing inflammatory pathway activity. The most selective candidate, DX1, demonstrated a significant reduction (87%) in transactivation compared to commercially available dexamethasone. DX1 also maintained 90% of the anti-inflammatory potential of dexamethasone while simultaneously displaying a reduced toxicity profile. Additionally, two novel and highly potent compounds, DX4 and PN4, were developed and shown to elicit similar mRNA expression at attomolar concentrations that dexamethasone exhibits at nanomolar dosages. To further explain these results, Molecular Dynamic (MD) simulations were performed to examine structural changes in the ligand-binding domain of the glucocorticoid receptor in response to docking with the top ligands. Differing interactions with the transcriptional activation function 2 (AF-2) region of the GR may be responsible for lower transactivation capacity in DX1. DX4 and PN4 lose contact with Arg611 due to a key interaction changing from a stronger hydrophilic to a weaker hydrophobic one, which leads to the formation of an unoccupied channel at the location of the deacylcortivazol (DAC)-expanded binding pocket. These findings provide insights into the structure-function relationships important for regulating anti-inflammatory activity, which has implications for clinical utility.
Collapse
Affiliation(s)
- Wesley B. Seaton
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA; (W.B.S.)
| | - Susan J. Burke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA (J.J.C.)
| | - Alexander R. Fisch
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA; (W.B.S.)
| | | | - Mary Grace A. Beck
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA (J.J.C.)
| | | | - Innocence Harvey
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA (J.J.C.)
| | - Molly S. Fontenot
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA (J.J.C.)
| | - J. Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA (J.J.C.)
| | - Shawn R. Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA; (W.B.S.)
| |
Collapse
|
6
|
Fang Y, Xiao C, Wang L, Wang Y, Zeng J, Liang Y, Huang R, Shi Y, Wu S, Du X, Sun S, Li M, Zheng Y, Wu H, Guo Q, Yang W. Synergistic Enhancement of Isoforskolin and Dexamethasone Against Sepsis and Acute Lung Injury Mouse Models. J Inflamm Res 2023; 16:5989-6001. [PMID: 38088941 PMCID: PMC10712681 DOI: 10.2147/jir.s421232] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 10/09/2023] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND Sepsis is initiated by the dysfunctional response of the host immune system to infection. Septic shock and acute lung injury (ALI) are the main etiology of death caused by sepsis. Glucocorticoids, which are commonly used in clinic to antagonize the inflammatory response of sepsis, may cause serious side effects. Isoforskolin (ISOF) from the plant Coleus forskohlii stimulates adenylyl cyclase, increases the cAMP level and inhibits inflammatory response. The aim of this study was to investigate the synergistic effect of ISOF with dexamethasone (DEX) to prevent and ameliorate septic inflammation. METHODS Lipopolysaccharide (LPS) of 30 and 5 mg/kg (iv.) was used to induce sepsis and ALI mice model respectively in vivo. BEAS-2B cells stimulated by LPS were applied as cell model in vitro. The cumulative survival of mice with LPS-induced sepsis and the histopathological changes of lungs in mice with acute lung injury were observed, and the secretion of pro-inflammatory cytokines was analyzed by ELISA. The expression of RGS2 in BEAS-2B cells was detected by immunoblotting assay and PCR. RESULTS In the sepsis mice model, ISOF (10 mg/kg) combined with DEX (10 mg/kg.) (ip.) pretreatment significantly increased mice survival rate from 33.3% to 58.3%, which was significantly higher than that of ISOF or DEX treated alone. In the ALI mice model, ISOF, DEX pretreatment alone and combined application attenuated pulmonary pathological changes in ALI mice. Furthermore, ISOF, DEX alone or combined administration decreased MPO, MDA, IL-6, and IL-8 levels, while significantly synergistic effects were observed in the combined treatment group compared with ISOF or DEX alone. In BEAS-2B cells, combined pretreatment with ISOF and DEX significantly decreased the expression of IL-8 and increased the expression of RGS2. CONCLUSION The results indicated that ISOF in combination with DEX synergistically improves survival rate and attenuates ALI in mice model through anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Yan Fang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Lueli Wang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Youlan Wang
- Kunming Institute of Medical Sciences, Kunming, People’s Republic of China
| | - Jun Zeng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Yaping Liang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Rong Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Yunke Shi
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Sha Wu
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Xiaohua Du
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shibo Sun
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Min Li
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Yuanyuan Zheng
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Hongxiang Wu
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Qiuzhe Guo
- Fuwai Yunnan Cardiovascular Hospital, Kunming, People’s Republic of China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
7
|
Chatziparasidis G, Bush A. Enigma variations: The multi-faceted problems of pre-school wheeze. Pediatr Pulmonol 2022; 57:1990-1997. [PMID: 35652262 DOI: 10.1002/ppul.26027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 11/08/2022]
Abstract
Numerous publications on wheezing disorders in children younger than 6 years have appeared in the medical literature over the last decades with the aim of shedding light on the mechanistic pathways (endotypes) and treatment. Nevertheless, there is yet no consensus as to the appropriate way to manage preschool wheeze mainly because of the lack of a clear definition of "preschool asthma" and the paucity of scientific evidence concerning its underlying endotypes. A symptom-based approach is inadequate since the human airway can respond to external stimuli with a limited range of symptoms and signs, including cough and wheeze, and these manifestations represent the final expression of many clinical entities with potentially different pathophysiologies requiring different individualized treatments. Hence, new studies challenge the symptom-based approach and promote the importance of managing the wheezy child based on the "airway phenotype." This will enable the clinician to identify not only the child with a serious underlying pathology (e.g., a structural airway disorder or immunodeficiency) who is in need of prompt and specific treatment but also increase the specificity of treatment for the child with symptoms suggestive of an "asthma" syndrome. In the latter case, focus should be given to the identification of treatable traits. This review summarizes the current understanding in management of preschool wheezing and highlights the unmet need for further research.
Collapse
Affiliation(s)
- Grigorios Chatziparasidis
- Department of Paediatrics, Metropolitan Hospital, Athens, and Primary Cilia Dyskinesia Unit, University of Thessaly, Volos, Greece
| | - Andrew Bush
- Departments of Paediatrics and Paediatric Respiratory Medicine, Royal Brompton Harefield NHS Foundation Trust and Imperial College, London, UK
| |
Collapse
|
8
|
Gebski EB, Anaspure O, Panettieri RA, Koziol-White CJ. Airway smooth muscle and airway hyperresponsiveness in asthma: mechanisms of airway smooth muscle dysfunction. Minerva Med 2022; 113:4-16. [PMID: 33496164 PMCID: PMC9254130 DOI: 10.23736/s0026-4806.21.07283-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Airway smooth muscle plays a pivotal role in modulating bronchomotor tone. Modulation of contractile and relaxation signaling is critical to alleviate the airway hyperresponsiveness (AHR) associated with asthma. Emerging studies examining the phenotype of ASM in the context of asthma provide rich avenues to develop more effective therapeutics to attenuate the AHR associated with the disease.
Collapse
Affiliation(s)
- Eric B Gebski
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Omkar Anaspure
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA -
| |
Collapse
|
9
|
Pera T, Loblundo C, Penn RB. Pharmacological Management of Asthma and COPD. COMPREHENSIVE PHARMACOLOGY 2022:762-802. [DOI: 10.1016/b978-0-12-820472-6.00095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Pelaia C, Crimi C, Crimi N, Ricciardi L, Scichilone N, Valenti G, Bonavita O, Andaloro S, Morini P, Rizzi A, Pelaia G. Indacaterol/glycopyrronium/mometasone fixed dose combination for uncontrolled asthma. Expert Rev Respir Med 2021; 16:183-195. [PMID: 34845963 DOI: 10.1080/17476348.2021.2011222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Asthma symptoms can be relieved through a maintenance treatment combining long-acting β2-agonist and inhaled corticosteroids (LABA/ICS). However, for patients with inadequately controlled asthma, the LABA/ICS combination might not be sufficient, and clinical guidelines recommend the administration of inhaled long-acting muscarinic antagonists (LAMA) as an add-on therapy to better control asthma and improve lung function. For nearly two decades, the only LAMA to be approved on the market has been tiotropium. AREAS COVERED We reviewed recent clinical studies evaluating the safety and efficacy of LABA/LAMA/ICS fixed dose combinations by searching the PubMed database. Molecular mechanisms and clinical data support the use of a once-daily, single-inhaler fixed dose combination of the LABA/LAMA/ICS indacaterol/glycopyrronium/mometasone (IND/GLY/MF), the first therapy combining three agents in a fixed dose approved in Europe for the treatment of uncontrolled asthma. EXPERT OPINION IND/GLY/MF was superior to both IND/MF and salmeterol/fluticasone, a well-established LABA/ICS combination improving the lung function in uncontrolled asthma. Moreover, IND/GLY/MF, delivered through the Breezhaler inhaler in a single inhalation, is the first inhaled therapy prescribed alongside a digital companion, a sensor and the Propeller app, allowing for improved treatment adherence, reduced rescue inhaler usage and hospitalizations, increased patient satisfaction and asthma control.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Claudia Crimi
- Respiratory Disease Unit, University Hospital of Catania, Catania, Italy
| | - Nunzio Crimi
- Respiratory Disease Unit, University Hospital of Catania, Catania, Italy
| | - Luisa Ricciardi
- Allergy and Clinical Immunology Unit, A.O.U. Policlinico "G. Martino", Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Nicola Scichilone
- Medicine of the Respiratory System, Hospital 'P. Giaccone', University of Palermo, Palermo, Italy
| | - Giuseppe Valenti
- Allergology and Pulmonology Unit, Pta Biondo, ASP Palermo, Italy
| | | | | | - Paolo Morini
- Medical Department, Novartis Farma Origgio, Varese, Italy
| | - Andrea Rizzi
- Medical Department, Novartis Farma Origgio, Varese, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
11
|
Berman R, Rose CS, Downey GP, Day BJ, Chu HW. Role of Particulate Matter from Afghanistan and Iraq in Deployment-Related Lung Disease. Chem Res Toxicol 2021; 34:2408-2423. [PMID: 34808040 DOI: 10.1021/acs.chemrestox.1c00090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Approximately 3 million United States military personnel and contractors were deployed to Southwest Asia and Afghanistan over the past two decades. After returning to the United States, many developed persistent respiratory symptoms, including those due to asthma, rhinosinusitis, bronchiolitis, and others, which we collectively refer to as deployment-related lung diseases (DRLD). The mechanisms of different DRLD have not been well defined. Limited studies from us and others suggest that multiple factors and biological signaling pathways contribute to the onset of DRLD. These include, but are not limited to, exposures to high levels of particulate matter (PM) from sandstorms, burn pit combustion products, improvised explosive devices, and diesel exhaust particles. Once inhaled, these hazardous substances can activate lung immune and structural cells to initiate numerous cell-signaling pathways such as oxidative stress, Toll-like receptors, and cytokine-driven cell injury (e.g., interleukin-33). These biological events may lead to a pro-inflammatory response and airway hyperresponsiveness. Additionally, exposures to PM and other environmental hazards may predispose military personnel and contractors to more severe disease due to the interactions of those hazardous materials with subsequent exposures to allergens and cigarette smoke. Understanding how airborne exposures during deployment contribute to DRLD may identify effective targets to alleviate respiratory diseases and improve quality of life in veterans and active duty military personnel.
Collapse
Affiliation(s)
- Reena Berman
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Cecile S Rose
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Gregory P Downey
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Brian J Day
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| |
Collapse
|
12
|
Kim Y, Hou V, Huff RD, Aguiar JA, Revill S, Tiessen N, Cao Q, Miller MS, Inman MD, Ask K, Doxey AC, Hirota JA. Potentiation of long-acting β 2-agonist and glucocorticoid responses in human airway epithelial cells by modulation of intracellular cAMP. Respir Res 2021; 22:266. [PMID: 34666750 PMCID: PMC8527633 DOI: 10.1186/s12931-021-01862-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/09/2021] [Indexed: 11/10/2022] Open
Abstract
Introduction Over 300 million people in the world live with asthma, resulting in 500,000 annual global deaths with future increases expected. It is estimated that around 50–80% of asthma exacerbations are due to viral infections. Currently, a combination of long-acting beta agonists (LABA) for bronchodilation and glucocorticoids (GCS) to control lung inflammation represent the dominant strategy for the management of asthma, however, it is still sub-optimal in 35–50% of moderate-severe asthmatics resulting in persistent lung inflammation, impairment of lung function, and risk of mortality. Mechanistically, LABA/GCS combination therapy results in synergistic efficacy mediated by intracellular cyclic adenosine monophosphate (cAMP). Hypothesis Increasing intracellular cAMP during LABA/GCS combination therapy via inhibiting phosphodiesterase 4 (PDE4) and/or blocking the export of cAMP by ATP Binding Cassette Transporter C4 (ABCC4), will potentiate anti-inflammatory responses of mainstay LABA/GCS therapy. Methods Expression and localization experiments were performed using in situ hybridization and immunohistochemistry in human lung tissue from healthy subjects, while confirmatory transcript and protein expression analyses were performed in primary human airway epithelial cells and cell lines. Intervention experiments were performed on the human airway epithelial cell line, HBEC-6KT, by pre-treatment with combinations of LABA/GCS with PDE4 and/or ABCC4 inhibitors followed by Poly I:C or imiquimod challenge as a model for viral stimuli. Cytokine readouts for IL-6, IL-8, CXCL10/IP-10, and CCL5/RANTES were quantified by ELISA. Results Using archived human lung and human airway epithelial cells, ABCC4 gene and protein expression were confirmed in vitro and in situ. LABA/GCS attenuation of Poly I:C or imiquimod-induced IL-6 and IL-8 were potentiated with ABCC4 and PDE4 inhibition, which was greater when ABCC4 and PDE4 inhibition was combined. Modulation of cAMP levels had no impact on LABA/GCS modulation of Poly I:C-induced CXCL10/IP-10 or CCL5/RANTES. Conclusion Modulation of intracellular cAMP levels by PDE4 or ABCC4 inhibition potentiates LABA/GCS efficacy in human airway epithelial cells challenged with viral stimuli. The data suggest further exploration of the value of adding cAMP modulators to mainstay LABA/GCS therapy in asthma for potentiated anti-inflammatory efficacy.
Collapse
Affiliation(s)
- Yechan Kim
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Vincent Hou
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Ryan D Huff
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, V6H 3Z, Canada
| | - Jennifer A Aguiar
- Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Spencer Revill
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Nicholas Tiessen
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Quynh Cao
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Matthew S Miller
- Department of Biochemistry, McMaster University, Hamilton, ON, L8S 4K1, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Mark D Inman
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Andrew C Doxey
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada.,Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Jeremy A Hirota
- Firestone Institute for Respiratory Health-Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada. .,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, V6H 3Z, Canada. .,Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada. .,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
13
|
Lu L, Huang J, Deng X, Sun X, Dong J. Application of glucocorticoids in patients with novel coronavirus infection: From bench to bedside. TRADITIONAL MEDICINE AND MODERN MEDICINE 2021. [DOI: 10.1142/s257590002030009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glucocorticoids (GCs) have potential anti-inflammatory and immunosuppressive effects. There is plenty of controversy about the application of glucocorticoids in the treatment of coronavirus disease 2019 (COVID-19). This paper briefly summarizes the mechanism of glucocorticoids and their receptors and clinical applications in COVID-19. Through reviewing the current literature, our aim is to have a deeper understanding of the mechanism of GCs and their clinical applications, so as to find possible ways to enhance their efficacy and reduce drug resistance or side effects.
Collapse
Affiliation(s)
- Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, P. R. China
- Qingpu Chinese Medicine Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, P. R. China
| | - Jianhua Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, P. R. China
- Qingpu Chinese Medicine Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, P. R. China
| | - Xiaohong Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, P. R. China
- Qingpu Chinese Medicine Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, P. R. China
| | - Xianjun Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, P. R. China
- Qingpu Chinese Medicine Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, P. R. China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, P. R. China
- Qingpu Chinese Medicine Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, P. R. China
| |
Collapse
|
14
|
Fuentes N, McCullough M, Panettieri RA, Druey KM. RGS proteins, GRKs, and beta-arrestins modulate G protein-mediated signaling pathways in asthma. Pharmacol Ther 2021; 223:107818. [PMID: 33600853 PMCID: PMC8192426 DOI: 10.1016/j.pharmthera.2021.107818] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Asthma is a highly prevalent disorder characterized by chronic lung inflammation and reversible airways obstruction. Pathophysiological features of asthma include episodic and reversible airway narrowing due to increased bronchial smooth muscle shortening in response to external and host-derived mediators, excessive mucus secretion into the airway lumen, and airway remodeling. The aberrant airway smooth muscle (ASM) phenotype observed in asthma manifests as increased sensitivity to contractile mediators (EC50) and an increase in the magnitude of contraction (Emax); collectively these attributes have been termed "airways hyper-responsiveness" (AHR). This defining feature of asthma can be promoted by environmental factors including airborne allergens, viruses, and air pollution and other irritants. AHR reduces airway caliber and obstructs airflow, evoking clinical symptoms such as cough, wheezing and shortness of breath. G-protein-coupled receptors (GPCRs) have a central function in asthma through their impact on ASM and airway inflammation. Many but not all treatments for asthma target GPCRs mediating ASM contraction or relaxation. Here we discuss the roles of specific GPCRs, G proteins, and their associated signaling pathways, in asthma, with an emphasis on endogenous mechanisms of GPCR regulation of ASM tone and lung inflammation including regulators of G-protein signaling (RGS) proteins, G-protein coupled receptor kinases (GRKs), and β-arrestin.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Morgan McCullough
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ, United States of America
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America.
| |
Collapse
|
15
|
Sharma P, Penn RB. Can GPCRs Be Targeted to Control Inflammation in Asthma? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:1-20. [PMID: 34019260 DOI: 10.1007/978-3-030-68748-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Historically, the drugs used to manage obstructive lung diseases (OLDs), asthma, and chronic obstructive pulmonary disease (COPD) either (1) directly regulate airway contraction by blocking or relaxing airway smooth muscle (ASM) contraction or (2) indirectly regulate ASM contraction by inhibiting the principal cause of ASM contraction/bronchoconstriction and airway inflammation. To date, these tasks have been respectively assigned to two diverse drug types: agonists/antagonists of G protein-coupled receptors (GPCRs) and inhaled or systemic steroids. These two types of drugs "stay in their lane" with respect to their actions and consequently require the addition of the other drug to effectively manage both inflammation and bronchoconstriction in OLDs. Indeed, it has been speculated that safety issues historically associated with beta-agonist use (beta-agonists activate the beta-2-adrenoceptor (β2AR) on airway smooth muscle (ASM) to provide bronchoprotection/bronchorelaxation) are a function of pro-inflammatory actions of β2AR agonism. Recently, however, previously unappreciated roles of various GPCRs on ASM contractility and on airway inflammation have been elucidated, raising the possibility that novel GPCR ligands targeting these GPCRs can be developed as anti-inflammatory therapeutics. Moreover, we now know that many GPCRs can be "tuned" and not just turned "off" or "on" to specifically activate the beneficial therapeutic signaling a receptor can transduce while avoiding detrimental signaling. Thus, the fledging field of biased agonism pharmacology has the potential to turn the β2AR into an anti-inflammatory facilitator in asthma, possibly reducing or eliminating the need for steroids.
Collapse
Affiliation(s)
- Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy, & Critical Care Medicine Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College Thomas Jefferson University, Philadelphia, PA, USA
| | - Raymond B Penn
- Center for Translational Medicine, Division of Pulmonary, Allergy, & Critical Care Medicine Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Abdullah K, Fell DB, Radhakrishnan D, Hawken S, Johnson DW, Mandhane P, To T, Joubert G, Plint AC. Risk of asthma in children diagnosed with bronchiolitis during infancy: protocol of a longitudinal cohort study linking emergency department-based clinical data to provincial health administrative databases. BMJ Open 2021; 11:e048823. [PMID: 33941638 PMCID: PMC8098926 DOI: 10.1136/bmjopen-2021-048823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION The Canadian Bronchiolitis Epinephrine Steroid Trial (CanBEST) and the Bronchiolitis Severity Cohort (BSC) study enrolled infants with bronchiolitis during the first year of life. The CanBEST trial suggested that treatment of infants with a combined therapy of high-dose corticosteroids and nebulised epinephrine reduced the risk of admission to hospital. Our study aims to-(1) quantify the risk of developing asthma by age 5 and 10 years in children treated with high-dose corticosteroid and epinephrine for bronchiolitis during infancy, (2) identify risk factors associated with development of asthma in children with bronchiolitis during infancy, (3) develop asthma prediction models for children diagnosed with bronchiolitis during infancy. METHODS AND ANALYSIS We propose a longitudinal cohort study in which we will link data from the CanBEST and BSC study with routinely collected data from provincial health administrative databases. Our outcome is asthma incidence measured using a validated health administrative data algorithm. Primary exposure will be treatment with a combined therapy of high-dose corticosteroids and nebulised epinephrine for bronchiolitis. Covariates will include type of viral pathogen, disease severity, medication use, maternal, prenatal, postnatal and demographic factors and variables related to health service utilisation for acute lower respiratory tract infection. The risk associated with development of asthma in children treated with high-dose corticosteroid and epinephrine for bronchiolitis will be assessed using multivariable Cox proportional hazards regression models. Prediction models will be developed using multivariable logistic regression analysis and internally validated using a bootstrap approach. ETHICS AND DISSEMINATION Our study has been approved by the ethics board of all four participating sites of the CanBEST and BSC study. Finding of the study will be disseminated to the academic community and relevant stakeholders through conferences and peer-reviewed publications. TRIAL REGISTRATION NUMBER ISRCTN56745572; Post-results.
Collapse
Affiliation(s)
- Kawsari Abdullah
- Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- ICES, Ottawa, Ontario, Canada
| | - Deshayne B Fell
- Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- ICES, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dhenuka Radhakrishnan
- Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- ICES, Ottawa, Ontario, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Steven Hawken
- ICES, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David W Johnson
- Departments of Pediatrics and Physiology and Pharmacology, University of Calgary, Calgery, Alberta, Canada
- Maternal Newborn Child & Youth SCN, Alberta Health Services, Calgery, Alberta, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Teresa To
- ICES, Ottawa, Ontario, Canada
- Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gary Joubert
- London Health Sciences Centre, London, Ontario, Canada
| | - Amy C Plint
- Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Mostafa MM, Rider CF, Wathugala ND, Leigh R, Giembycz MA, Newton R. Transcriptome-Level Interactions between Budesonide and Formoterol Provide Insight into the Mechanism of Action of Inhaled Corticosteroid/Long-Acting β 2-Adrenoceptor Agonist Combination Therapy in Asthma. Mol Pharmacol 2021; 99:197-216. [PMID: 33376135 DOI: 10.1124/molpharm.120.000146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/07/2020] [Indexed: 12/26/2022] Open
Abstract
In 2019, the Global Initiative for Asthma treatment guidelines were updated to recommend that inhaled corticosteroid (ICS)/long-acting β 2-adrenoceptor agonist (LABA) combination therapy should be a first-in-line treatment option for asthma. Although clinically superior to ICS, mechanisms underlying the efficacy of this combination therapy remain unclear. We hypothesized the existence of transcriptomic interactions, an effect that was tested in BEAS-2B and primary human bronchial epithelial cells (pHBECs) using formoterol and budesonide as representative LABA and ICS, respectively. In BEAS-2B cells, formoterol produced 267 (212 induced; 55 repressed) gene expression changes (≥2/≤0.5-fold) that were dominated by rapidly (1 to 2 hours) upregulated transcripts. Conversely, budesonide induced 370 and repressed 413 mRNAs, which occurred predominantly at 6-18 hours and was preceded by transcripts enriched in transcriptional regulators. Significantly, genes regulated by both formoterol and budesonide were over-represented in the genome; moreover, budesonide plus formoterol induced and repressed 609 and 577 mRNAs, respectively, of which ∼one-third failed the cutoff criterion for either treatment alone. Although induction of many mRNAs by budesonide plus formoterol was supra-additive, the dominant (and potentially beneficial) effect of budesonide on formoterol-induced transcripts, including those encoding many proinflammatory proteins, was repression. Gene ontology analysis of the budesonide-modulated transcriptome returned enriched terms for transcription, apoptosis, proliferation, differentiation, development, and migration. This "functional" ICS signature was augmented in the presence of formoterol. Thus, LABAs modulate glucocorticoid action, and comparable transcriptome-wide interactions in pHBECs imply that such effects may be extrapolated to individuals with asthma taking combination therapy. Although repression of formoterol-induced proinflammatory mRNAs should be beneficial, the pathophysiological consequences of other interactions require investigation. SIGNIFICANCE STATEMENT: In human bronchial epithelial cells, formoterol, a long-acting β 2-adrenoceptor agonist (LABA), enhanced the expression of inflammatory genes, and many of these changes were reduced by the glucocorticoid budesonide. Conversely, the ability of formoterol to enhance both gene induction and repression by budesonide provides mechanistic insight as to how adding a LABA to an inhaled corticosteroid may improve clinical outcomes in asthma.
Collapse
Affiliation(s)
- Mahmoud M Mostafa
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Christopher F Rider
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - N Dulmini Wathugala
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Richard Leigh
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
18
|
Joshi R, Hamed O, Yan D, Michi AN, Mostafa MM, Wiehler S, Newton R, Giembycz MA. Prostanoid Receptors of the EP 4-Subtype Mediate Gene Expression Changes in Human Airway Epithelial Cells with Potential Anti-Inflammatory Activity. J Pharmacol Exp Ther 2021; 376:161-180. [PMID: 33158942 DOI: 10.1124/jpet.120.000196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022] Open
Abstract
There is a clear, unmet clinical need to identify new drugs to treat individuals with asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF) in whom current medications are either inactive or suboptimal. In preclinical models, EP4-receptor agonists display efficacy, but their mechanism of action is unclear. In this study, using human bronchial epithelial cells as a therapeutically relevant drug target, we hypothesized that changes in gene expression may play an important role. Several prostanoid receptor mRNAs were detected in BEAS-2B cells, human primary bronchial epithelial cells (HBECs) grown in submersion culture and HBECs grown at an air-liquid interface with PTGER4 predominating. By using the activation of a cAMP response element reporter in BEAS-2B cells as a surrogate of gene expression, Schild analysis determined that PTGER4 mRNAs encoded functional EP4-receptors. Moreover, inhibitors of phosphodiesterase 4 (roflumilast N-oxide [RNO]) and cAMP-dependent protein kinase augmented and attenuated, respectively, reporter activation induced by 2-[3-[(1R,2S,3R)-3-hydroxy-2-[(E,3S)-3-hydroxy-5-[2-(methoxymethyl)phenyl]pent-1-enyl]-5-oxo-cyclopentyl]sulphanylpropylsulphanyl] acetic acid (ONO-AE1-329), a selective EP4-receptor agonist. ONO-AE1-329 also enhanced dexamethasone-induced activation of a glucocorticoid response element reporter in BEAS-2B cells, which was similarly potentiated by RNO. In each airway epithelial cell variant, numerous genes that may impart therapeutic benefit in asthma, COPD, and/or IPF were differentially expressed by ONO-AE1-329, and those changes were often augmented by RNO and/or dexamethasone. We submit that an EP4-receptor agonist, either alone or as a combination therapy, may be beneficial in individuals with chronic lung diseases in whom current treatment options are inadequate. SIGNIFICANCE STATEMENT: Using human bronchial epithelial cells as a therapeutically relevant drug target, we report that EP4-receptor activation promoted gene expression changes that could provide therapeutic benefit in individuals with asthma, COPD, and IPF in whom current treatment options are ineffective or suboptimal.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Omar Hamed
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dong Yan
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aubrey N Michi
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mahmoud M Mostafa
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shahina Wiehler
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Nguyen JP, Kim Y, Cao Q, Hirota JA. Interactions between ABCC4/MRP4 and ABCC7/CFTR in human airway epithelial cells in lung health and disease. Int J Biochem Cell Biol 2021; 133:105936. [PMID: 33529712 DOI: 10.1016/j.biocel.2021.105936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022]
Abstract
ATP binding cassette (ABC) transporters are present in all three domains of life - Archaea, Bacteria, and Eukarya. The conserved nature is a testament to the importance of these transporters in regulating endogenous and exogenous substrates required for life to exist. In humans, 49 ABC transporters have been identified to date with broad expression in different lung cell types with multiple transporter family members contributing to lung health and disease. The ABC transporter most commonly known to be linked to lung pathology is ABCC7, also known as cystic fibrosis transmembrane conductance regulator - CFTR. Closely related to the CFTR genomic sequence is ABCC4/multi-drug resistance protein-4. Genomic proximity is shared with physical proximity, with ABCC4 and CFTR physically coupled in cell membrane microenvironments of epithelial cells to orchestrate functional consequences of cyclic-adenosine monophosphate (cAMP)-dependent second messenger signaling and extracellular transport of endogenous and exogenous substrates. The present concise review summarizes the emerging data defining a role of the (ABCC7/CFTR)-ABCC4 macromolecular complex in human airway epithelial cells as a physiologically important pathway capable of impacting endogenous and exogenous mediator transport and ion transport in both lung health and disease.
Collapse
Affiliation(s)
- Jenny P Nguyen
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Yechan Kim
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Quynh Cao
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Jeremy A Hirota
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada; McMaster Immunology Research Centre, McMaster University, Canada; Department of Biology, University of Waterloo, Canada; Department of Medicine, University of British Columbia, Canada.
| |
Collapse
|
20
|
Rogliani P, Ritondo BL, Cavalli F, Giorgino F, Girolami A, Pane G, Pezzuto G, Zerillo B, Puxeddu E, Ora J. Synergy across the drugs approved for the treatment of asthma. Minerva Med 2021; 113:17-30. [PMID: 33496162 DOI: 10.23736/s0026-4806.21.07266-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Inhaled corticosteroids are the cornerstone for the treatment of stable asthma, however, when disease severity increases, escalating therapy to combinations of drugs acting on distinct signalling pathways is required. It is advantageous to providing evidence of a synergistic interaction across drug combinations, as it allows optimizing bronchodilation while lowering the dose of single agents. In the respiratory pharmacology field, two statistical models are accepted as gold standard to characterize drug interactions, namely the Bliss Independence criterion and the Unified Theory. In this review, pharmacological interactions across drugs approved for the treatment of asthma have been systematically assessed. EVIDENCE ACQUISITION A comprehensive literature search was performed in MEDLINE for studies that used a validated pharmacological method for assessing drug interaction. The results were extracted and reported via qualitative synthesis. EVIDENCE SYNTHESIS Overall, 45 studies were identified from literature search and 5 met the inclusion criteria. Current evidence coming from ex vivo models of asthma indicates that drug combinations modulating bronchial contractility induce a synergistic bronchorelaxant effect. In murine models of lung inflammation, the combination between inhaled corticosteroids and β2- adrenoceptor agonists synergistically improve lung function and the inflammatory profile. CONCLUSIONS There is still limited knowledge regarding the mechanistic basis underlying pharmacological interactions across drugs approved for asthma. The synergism elicited by combined agents is an effect of class. Specifically designed clinical trials are needed to confirm the results coming from preclinical evidence, but also to establish the minimal dose for combined agents to induce a synergistic interaction and maximize bronchodilation.
Collapse
Affiliation(s)
- Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy - .,Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy -
| | - Beatrice L Ritondo
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Cavalli
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Federica Giorgino
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Andrea Girolami
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Gloria Pane
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Gabriella Pezzuto
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Bartolomeo Zerillo
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Ermanno Puxeddu
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
21
|
McNabb HJ, Gonzalez S, Muli CS, Sjögren B. N-Terminal Targeting of Regulator of G Protein Signaling Protein 2 for F-Box Only Protein 44-Mediated Proteasomal Degradation. Mol Pharmacol 2020; 98:677-685. [PMID: 33008920 DOI: 10.1124/molpharm.120.000061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/17/2020] [Indexed: 12/28/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins are negative modulators of G protein signaling that have emerged as promising drug targets to improve specificity and reduce side effects of G protein-coupled receptor-related therapies. Several small molecule RGS protein inhibitors have been identified; however, enhancing RGS protein function is often more clinically desirable but presents a challenge. Low protein levels of RGS2 are associated with various pathologies, including hypertension and heart failure. For this reason, RGS2 is a prominent example wherein enhancing its function would be beneficial. RGS2 is rapidly ubiquitinated and proteasomally degraded, providing a point of intervention for small molecule RGS2-stabilizing compounds. We previously identified a novel cullin-RING E3 ligase utilizing F-box only protein 44 (FBXO44) as the substrate recognition component. Here, we demonstrate that RGS2 associates with FBXO44 through a stretch of residues in its N terminus. RGS2 contains four methionine residues close to the N terminus that can act as alternative translation initiation sites. The shorter translation initiation variants display reduced ubiquitination and proteasomal degradation as a result of lost association with FBXO44. In addition, we show that phosphorylation of Ser3 may be an additional mechanism to protect RGS2 from FBXO44-mediated proteasomal degradation. These findings contribute to elucidating mechanisms regulating steady state levels of RGS2 protein and will inform future studies to develop small molecule RGS2 stabilizers. These would serve as novel leads in pathologies associated with low RGS2 protein levels, such as hypertension, heart failure, and anxiety. SIGNIFICANCE STATEMENT: E3 ligases provide a novel point of intervention for therapeutic development, but progress is hindered by the lack of available information about specific E3-substrate pairs. Here, we provide molecular detail on the recognition of regulator of G protein signaling protein 2 (RGS2) by its E3 ligase, increasing the potential for rational design of small molecule RGS2 protein stabilizers. These would be clinically useful in pathologies associated with low RGS2 protein levels, such as hypertension, heart failure, and anxiety.
Collapse
Affiliation(s)
- Harrison J McNabb
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Stephanie Gonzalez
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Christine S Muli
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Benita Sjögren
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| |
Collapse
|
22
|
McNabb HJ, Zhang Q, Sjögren B. Emerging Roles for Regulator of G Protein Signaling 2 in (Patho)physiology. Mol Pharmacol 2020; 98:751-760. [PMID: 32973086 DOI: 10.1124/molpharm.120.000111] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022] Open
Abstract
Since their discovery in the mid-1990s, regulator of G protein signaling (RGS) proteins have emerged as key regulators of signaling through G protein-coupled receptors. Among the over 20 known RGS proteins, RGS2 has received increasing interest as a potential therapeutic drug target with broad clinical implications. RGS2 is a member of the R4 subfamily of RGS proteins and is unique in that it is selective for Gα q Despite only having an RGS domain, responsible for the canonical GTPase activating protein activity, RGS2 can regulate additional processes, such as protein synthesis and adenylate cyclase activity, through protein-protein interactions. Here we provide an update of the current knowledge of RGS2 function as it relates to molecular mechanisms of regulation as well as its potential role in regulating a number of physiologic systems and pathologies, including cardiovascular disease and central nervous system disorders, as well as various forms of cancer. SIGNIFICANCE STATEMENT: Regulator of G protein signaling (RGS) proteins represent an exciting class of novel drug targets. RGS2, in particular, could have broad clinical importance. As more details are emerging on the regulation of RGS2 in various physiological systems, the potential utility of this small protein in therapeutic development is increasing.
Collapse
Affiliation(s)
- Harrison J McNabb
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Qian Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Benita Sjögren
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| |
Collapse
|
23
|
Wang S, Xie Y, Huo YW, Li Y, Abel PW, Jiang H, Zou X, Jiao HZ, Kuang X, Wolff DW, Huang YG, Casale TB, Panettieri RA, Wei T, Cao Z, Tu Y. Airway relaxation mechanisms and structural basis of osthole for improving lung function in asthma. Sci Signal 2020; 13:eaax0273. [PMID: 33234690 PMCID: PMC8720283 DOI: 10.1126/scisignal.aax0273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Overuse of β2-adrenoceptor agonist bronchodilators evokes receptor desensitization, decreased efficacy, and an increased risk of death in asthma patients. Bronchodilators that do not target β2-adrenoceptors represent a critical unmet need for asthma management. Here, we characterize the utility of osthole, a coumarin derived from a traditional Chinese medicine, in preclinical models of asthma. In mouse precision-cut lung slices, osthole relaxed preconstricted airways, irrespective of β2-adrenoceptor desensitization. Osthole administered in murine asthma models attenuated airway hyperresponsiveness, a hallmark of asthma. Osthole inhibited phosphodiesterase 4D (PDE4D) activity to amplify autocrine prostaglandin E2 signaling in airway smooth muscle cells that eventually triggered cAMP/PKA-dependent relaxation of airways. The crystal structure of the PDE4D complexed with osthole revealed that osthole bound to the catalytic site to prevent cAMP binding and hydrolysis. Together, our studies elucidate a specific molecular target and mechanism by which osthole induces airway relaxation. Identification of osthole binding sites on PDE4D will guide further development of bronchodilators that are not subject to tachyphylaxis and would thus avoid β2-adrenoceptor agonist resistance.
Collapse
Affiliation(s)
- Sheng Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Xie
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yan-Wu Huo
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Peter W Abel
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Haihong Jiang
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Xiaohan Zou
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hai-Zhan Jiao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolin Kuang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dennis W Wolff
- Kansas City University of Medicine and Biosciences-Joplin, Joplin, MO 64804, USA
| | - You-Guo Huang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Thomas B Casale
- Department of Internal Medicine, University of South Florida School of Medicine, Tampa, FL 33612, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Taotao Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Yaping Tu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
24
|
Wong GS, Redes JL, Balenga N, McCullough M, Fuentes N, Gokhale A, Koziol-White C, Jude JA, Madigan LA, Chan EC, Jester WH, Biardel S, Flamand N, Panettieri RA, Druey KM. RGS4 promotes allergen- and aspirin-associated airway hyperresponsiveness by inhibiting PGE2 biosynthesis. J Allergy Clin Immunol 2020; 146:1152-1164.e13. [PMID: 32199913 PMCID: PMC7501178 DOI: 10.1016/j.jaci.2020.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 02/21/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Allergens elicit host production of mediators acting on G-protein-coupled receptors to regulate airway tone. Among these is prostaglandin E2 (PGE2), which, in addition to its role as a bronchodilator, has anti-inflammatory actions. Some patients with asthma develop bronchospasm after the ingestion of aspirin and other nonsteroidal anti-inflammatory drugs, a disorder termed aspirin-exacerbated respiratory disease. This condition may result in part from abnormal dependence on the bronchoprotective actions of PGE2. OBJECTIVE We sought to understand the functions of regulator of G protein signaling 4 (RGS4), a cytoplasmic protein expressed in airway smooth muscle and bronchial epithelium that regulates the activity of G-protein-coupled receptors, in asthma. METHODS We examined RGS4 expression in human lung biopsies by immunohistochemistry. We assessed airways hyperresponsiveness (AHR) and lung inflammation in germline and airway smooth muscle-specific Rgs4-/- mice and in mice treated with an RGS4 antagonist after challenge with Aspergillus fumigatus. We examined the role of RGS4 in nonsteroidal anti-inflammatory drug-associated bronchoconstriction by challenging aspirin-exacerbated respiratory disease-like (ptges1-/-) mice with aspirin. RESULTS RGS4 expression in respiratory epithelium is increased in subjects with severe asthma. Allergen-induced AHR was unexpectedly diminished in Rgs4-/- mice, a finding associated with increased airway PGE2 levels. RGS4 modulated allergen-induced PGE2 secretion in human bronchial epithelial cells and prostanoid-dependent bronchodilation. The RGS4 antagonist CCG203769 attenuated AHR induced by allergen or aspirin challenge of wild-type or ptges1-/- mice, respectively, in association with increased airway PGE2 levels. CONCLUSIONS RGS4 may contribute to the development of AHR by reducing airway PGE2 biosynthesis in allergen- and aspirin-induced asthma.
Collapse
Affiliation(s)
- Gordon S Wong
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Jamie L Redes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Nariman Balenga
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Morgan McCullough
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Nathalie Fuentes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Ameya Gokhale
- Food Allergy Research Unit, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, Md
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ
| | - Joseph A Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ
| | - Laura A Madigan
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - Eunice C Chan
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md
| | - William H Jester
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ
| | - Sabrina Biardel
- Centre de recherche de l'IUCPQ, Département de médecine, Faculté de médecine, Université Laval, Québec, Canada
| | - Nicolas Flamand
- Centre de recherche de l'IUCPQ, Département de médecine, Faculté de médecine, Université Laval, Québec, Canada
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), Bethesda, Md.
| |
Collapse
|
25
|
Berman R, Kopf KW, Min E, Huang J, Downey GP, Alam R, Chu HW, Day BJ. IL-33/ST2 signaling modulates Afghanistan particulate matter induced airway hyperresponsiveness in mice. Toxicol Appl Pharmacol 2020; 404:115186. [PMID: 32777237 DOI: 10.1016/j.taap.2020.115186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022]
Abstract
Increased symptoms of asthma-like respiratory illnesses have been reported in soldiers returning from tours of duty in Afghanistan. Inhalation of desert particulate matter (PM) may contribute to this deployment-related lung disease (DRLD), but little is known about disease mechanisms. The IL-33 signaling pathway, including its receptor ST2, has been implicated in the pathogenesis of lung diseases including asthma, but its role in PM-mediated airway dysfunction has not been studied. The goal of this study was to investigate whether IL-33/ST2 signaling contributes to airway dysfunction in preclinical models of lung exposure to Afghanistan PM (APM). Wild-type (WT) and ST2 knockout (KO) mice on the BALB/C background were oropharyngeally instilled with a single dose of saline or 50 μg of APM in saline. Airway hyperresponsiveness (AHR) and inflammation were assessed after 24 h. In WT mice, a single APM exposure induced AHR and neutrophilic inflammation. Unlike the WT mice, ST2 KO mice that lack the receptor for IL-33 did not demonstrate AHR although airway neutrophilic inflammation was comparable to the WT mice. Oropharyngeal delivery of a soluble ST2 decoy receptor in APM-exposed WT mice significantly blocked AHR. Additional data in mouse tracheal epithelial cell and lung macrophage cultures demonstrated a role of APM-induced IL-33/ST2 signaling in suppression of regulator of G protein signaling 2 (RGS2), a gene known to protect against bronchoconstriction. We present for the first time that APM may increase AHR, one of the features of asthma, in part through the IL-33/ST2/RGS2 pathway.
Collapse
Affiliation(s)
- Reena Berman
- Department of Medicine, Basic Science Section, National Jewish Health, Denver, CO, United States of America
| | - Katrina W Kopf
- Biological Resource Center, National Jewish Health, Denver, CO, United States of America
| | - Elysia Min
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America
| | - Jie Huang
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America
| | - Gregory P Downey
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, CO, United States of America
| | - Rafeul Alam
- Department of Medicine, Division of Allergy & Clinical Immunology, National Jewish Health, Denver, CO, United States of America
| | - Hong Wei Chu
- Department of Medicine, Basic Science Section, National Jewish Health, Denver, CO, United States of America.
| | - Brian J Day
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America.
| |
Collapse
|
26
|
Chinn AM, Insel PA. Cyclic AMP in dendritic cells: A novel potential target for disease-modifying agents in asthma and other allergic disorders. Br J Pharmacol 2020; 177:3363-3377. [PMID: 32372523 DOI: 10.1111/bph.15095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/27/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Allergic diseases are immune disorders that are a global health problem, affecting a large portion of the world's population. Allergic asthma is a heterogeneous disease that alters the biology of the airway. A substantial portion of patients with asthma do not respond to conventional therapies; thus, new and effective therapeutics are needed. Dendritic cells (DCs), antigen presenting cells that regulate helper T cell differentiation, are key drivers of allergic inflammation but are not the target of current therapies. Here we review the role of dendritic cells in allergic conditions and propose a disease-modifying strategy for treating allergic asthma: cAMP-mediated inhibition of dendritic cells to blunt allergic inflammation. This approach contrasts with current treatments that focus on treating clinical manifestations of airway inflammation. Disease-modifying agents that target cAMP and its signalling pathway in dendritic cells may provide a novel means to treat asthma and other allergic diseases.
Collapse
Affiliation(s)
- Amy M Chinn
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
Anthracopoulos MB, Everard ML. Asthma: A Loss of Post-natal Homeostatic Control of Airways Smooth Muscle With Regression Toward a Pre-natal State. Front Pediatr 2020; 8:95. [PMID: 32373557 PMCID: PMC7176812 DOI: 10.3389/fped.2020.00095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
The defining feature of asthma is loss of normal post-natal homeostatic control of airways smooth muscle (ASM). This is the key feature that distinguishes asthma from all other forms of respiratory disease. Failure to focus on impaired ASM homeostasis largely explains our failure to find a cure and contributes to the widespread excessive morbidity associated with the condition despite the presence of effective therapies. The mechanisms responsible for destabilizing the normal tight control of ASM and hence airways caliber in post-natal life are unknown but it is clear that atopic inflammation is neither necessary nor sufficient. Loss of homeostasis results in excessive ASM contraction which, in those with poor control, is manifest by variations in airflow resistance over short periods of time. During viral exacerbations, the ability to respond to bronchodilators is partially or almost completely lost, resulting in ASM being "locked down" in a contracted state. Corticosteroids appear to restore normal or near normal homeostasis in those with poor control and restore bronchodilator responsiveness during exacerbations. The mechanism of action of corticosteroids is unknown and the assumption that their action is solely due to "anti-inflammatory" effects needs to be challenged. ASM, in evolutionary terms, dates to the earliest land dwelling creatures that required muscle to empty primitive lungs. ASM appears very early in embryonic development and active peristalsis is essential for the formation of the lungs. However, in post-natal life its only role appears to be to maintain airways in a configuration that minimizes resistance to airflow and dead space. In health, significant constriction is actively prevented, presumably through classic negative feedback loops. Disruption of this robust homeostatic control can develop at any age and results in asthma. In order to develop a cure, we need to move from our current focus on immunology and inflammatory pathways to work that will lead to an understanding of the mechanisms that contribute to ASM stability in health and how this is disrupted to cause asthma. This requires a radical change in the focus of most of "asthma research."
Collapse
Affiliation(s)
| | - Mark L. Everard
- Division of Paediatrics & Child Health, Perth Children's Hospital, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
28
|
Bouvet GF, Voisin G, Cyr Y, Bascunana V, Massé C, Berthiaume Y. DNA Methylation Regulates RGS2-induced S100A12 Expression in Airway Epithelial Cells. Am J Respir Cell Mol Biol 2019; 59:601-613. [PMID: 29944393 DOI: 10.1165/rcmb.2016-0164oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
RGS2 is a key modulator of stress in human airway epithelial cells, especially of hyperresponsiveness and mucin hypersecretion, both of which are features of cystic fibrosis (CF). Because its expression can be modulated through the DNA methylation pathway, we hypothesize that RGS2 is downregulated by DNA hypermethylation in CF airway epithelial cells. This downregulation would then lead to an enhanced inflammatory response. We demonstrated RGS2 transcript and protein downregulation in cultured airway epithelial cells from patients with CF and validated our findings in two CF epithelial cell lines. A methylated DNA immunoprecipitation array showed the presence of methylated cytosine on 13 gene promoters in CF. Among these genes, we confirmed that the RGS2 promoter was hypermethylated by using bisulfite conversion coupled with a methylation-specific PCR assay. Finally, we showed that downregulation of RGS2 in non-CF cells increased the expression of S100A12, a proinflammatory marker. These results highlight the importance of epigenetic regulation in gene expression in CF and show that RGS2 might modulate the inflammatory response in CF through DNA methylation control.
Collapse
Affiliation(s)
| | - Gregory Voisin
- Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| | - Yannick Cyr
- Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| | | | - Chantal Massé
- Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| | - Yves Berthiaume
- Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| |
Collapse
|
29
|
Mostafa MM, Rider CF, Shah S, Traves SL, Gordon PMK, Miller-Larsson A, Leigh R, Newton R. Glucocorticoid-driven transcriptomes in human airway epithelial cells: commonalities, differences and functional insight from cell lines and primary cells. BMC Med Genomics 2019; 12:29. [PMID: 30704470 PMCID: PMC6357449 DOI: 10.1186/s12920-018-0467-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Background Glucocorticoids act on the glucocorticoid receptor (GR; NR3C1) to resolve inflammation and, as inhaled corticosteroids (ICS), are the cornerstone of treatment for asthma. However, reduced efficacy in severe disease or exacerbations indicates a need to improve ICS actions. Methods Glucocorticoid-driven transcriptomes were compared using PrimeView microarrays between primary human bronchial epithelial (HBE) cells and the model cell lines, pulmonary type II A549 and bronchial epithelial BEAS-2B cells. Results In BEAS-2B cells, budesonide induced (≥2-fold, P ≤ 0.05) or, in a more delayed fashion, repressed (≤0.5-fold, P ≤ 0.05) the expression of 63, 133, 240, and 257 or 15, 56, 236, and 344 mRNAs at 1, 2, 6, and 18 h, respectively. Within the early-induced mRNAs were multiple transcriptional activators and repressors, thereby providing mechanisms for the subsequent modulation of gene expression. Using the above criteria, 17 (BCL6, BIRC3, CEBPD, ERRFI1, FBXL16, FKBP5, GADD45B, IRS2, KLF9, PDK4, PER1, RGCC, RGS2, SEC14L2, SLC16A12, TFCP2L1, TSC22D3) induced and 8 (ARL4C, FLRT2, IER3, IL11, PLAUR, SEMA3A, SLC4A7, SOX9) repressed mRNAs were common between A549, BEAS-2B and HBE cells at 6 h. As absolute gene expression change showed greater commonality, lowering the cut-off (≥1.25 or ≤ 0.8-fold) within these groups produced 93 induced and 82 repressed genes in common. Since large changes in few mRNAs and/or small changes in many mRNAs may drive function, gene ontology (GO)/pathway analyses were performed using both stringency criteria. Budesonide-induced genes showed GO term enrichment for positive and negative regulation of transcription, signaling, proliferation, apoptosis, and movement, as well as FOXO and PI3K-Akt signaling pathways. Repressed genes were enriched for inflammatory signaling pathways (TNF, NF-κB) and GO terms for cytokine activity, chemotaxis and cell signaling. Reduced growth factor expression and effects on proliferation and apoptosis were highlighted. Conclusions While glucocorticoids repress mRNAs associated with inflammation, prior induction of transcriptional activators and repressors may explain longer-term responses to these agents. Furthermore, positive and negative effects on signaling, proliferation, migration and apoptosis were revealed. Since many such gene expression changes occurred in human airways post-ICS inhalation, the effects observed in cell lines and primary HBE cells in vitro may be relevant to ICS in vivo. Electronic supplementary material The online version of this article (10.1186/s12920-018-0467-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mahmoud M Mostafa
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Cardiovascular and Respiratory Sciences graduate program, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Christopher F Rider
- Department of Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suharsh Shah
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Suzanne L Traves
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Paul M K Gordon
- Centre for Health Genomics and Informatics, University of Calgary, Calgary, Alberta, Canada
| | | | - Richard Leigh
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
30
|
Milara J, Díaz-Platas L, Contreras S, Ribera P, Roger I, Ballester B, Montero P, Cogolludo Á, Morcillo E, Cortijo J. MUC1 deficiency mediates corticosteroid resistance in chronic obstructive pulmonary disease. Respir Res 2018; 19:226. [PMID: 30458870 PMCID: PMC6247701 DOI: 10.1186/s12931-018-0927-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/01/2018] [Indexed: 01/10/2023] Open
Abstract
Background Lung inflammation in COPD is poorly controlled by inhaled corticosteroids (ICS). Strategies to improve ICS efficacy or the search of biomarkers who may select those patients candidates to receive ICS in COPD are needed. Recent data indicate that MUC1 cytoplasmic tail (CT) membrane mucin can mediate corticosteroid efficacy in chronic rhinosinusitis. The objective of this work was to analyze the previously unexplored role of MUC1 on corticosteroid efficacy in COPD in vitro and in vivo models. Methods MUC1-CT expression was measured by real time PCR, western blot, immunohistochemistry and immunofluorescence. The inflammatory mediators IL-8, MMP9, GM-CSF and MIP3α were measured by ELISA. The effect of MUC1 on inflammation and corticosteroid anti-inflammatory effects was measured using cell siRNA in vitro and Muc1-KO in vivo animal models. Results MUC1-CT expression was downregulated in lung tissue, bronchial epithelial cells and lung neutrophils from smokers (n = 11) and COPD (n = 11) patients compared with healthy subjects (n = 10). MUC1 was correlated with FEV1% (ρ = 0.7479; p < 0.0001) in smokers and COPD patients. Cigarette smoke extract (CSE) decreased the expression of MUC1 and induced corticosteroid resistance in human primary bronchial epithelial cells and human neutrophils. MUC1 Gene silencing using siRNA-MUC1 impaired the anti-inflammatory effects of dexamethasone and reduced glucocorticoid response element activation. Dexamethasone promoted glucocorticoid receptor alpha (GRα) and MUC1-CT nuclear translocation and co-localization that was inhibited by CSE. Lung function decline and inflammation induced by lipopolysaccharide and cigarette smoke in Muc1 KO mice was resistant to dexamethasone. Conclusions These results confirm a role for MUC1-CT mediating corticosteroid efficacy in COPD. Electronic supplementary material The online version of this article (10.1186/s12931-018-0927-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, Jaume I University, Castellón de la Plana, Spain. .,Pharmacy Unit, University General Hospital Consortium, Valencia, Spain. .,CIBERES, Health Institute Carlos III, Valencia, Spain. .,Unidad de Investigación Clínica, Consorcio Hospital General Universitario, Avenida tres cruces s/n, E-46014, Valencia, Spain.
| | - Lucía Díaz-Platas
- Unidade Radiofármacos PET, GALARIA, Santiago de Compostela, A Coruña, Spain
| | - Sonia Contreras
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Pilar Ribera
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Inés Roger
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Ángel Cogolludo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Esteban Morcillo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,Health Research Institute INCLIVA, Valencia, Spain
| | - Julio Cortijo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,Research and teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
31
|
George T, Chakraborty M, Giembycz MA, Newton R. A bronchoprotective role for Rgs2 in a murine model of lipopolysaccharide-induced airways inflammation. Allergy Asthma Clin Immunol 2018; 14:40. [PMID: 30305828 PMCID: PMC6166284 DOI: 10.1186/s13223-018-0266-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Asthma exacerbations are associated with the recruitment of neutrophils to the lungs. These cells release proteases and mediators, many of which act at G protein-coupled receptors (GPCRs) that couple via Gq to promote bronchoconstriction and inflammation. Common asthma therapeutics up-regulate expression of the regulator of G protein signalling (RGS), RGS2. As RGS2 reduces signaling from Gq-coupled GPCRs, we have defined role(s) for this GTPase-activating protein in an acute neutrophilic model of lung inflammation. Methods Wild type and Rgs2−/− C57Bl6 mice were exposed to nebulized lipopolysaccharide (LPS). Lung function (respiratory system resistance and compliance) was measured using a SCIREQ flexivent small animal ventilator. Lung inflammation was assessed by histochemistry, cell counting and by cytokine and chemokine expression in bronchoalveolar lavage (BAL) fluid. Results Lipopolysaccharide inhalation induced transient airways hyperreactivity (AHR) and neutrophilic lung inflammation. While AHR and inflammation was greatest 3 h post-LPS exposure, BAL neutrophils persisted for 24 h. At 3 h post-LPS inhalation, multiple inflammatory cytokines (CSF2, CSF3, IL6, TNF) and chemokines (CCL3, CCL4, CXCL1, CXCL2) were highly expressed in the BAL fluid, prior to declining by 24 h. Compared to wild type counterparts, Rgs2−/− mice developed significantly greater airflow resistance in response to inhaled methacholine (MCh) at 3 h post-LPS exposure. At 24 h post-LPS exposure, when lung function was recovering in the wild type animals, MCh-induced resistance was increased, and compliance decreased, in Rgs2−/− mice. Thus, Rgs2−/− mice show AHR and stiffer lungs 24 h post-LPS exposure. Histological markers of inflammation, total and differential cell counts, and major cytokine and chemokine expression in BAL fluid were similar between wild type and Rgs2−/− mice. However, 3 and 24 h post-LPS exposure, IL12B expression was significantly elevated in BAL fluid from Rgs2−/− mice compared to wild type animals. Conclusions While Rgs2 is bronchoprotective in acute neutrophilic inflammation, no clear anti-inflammatory effect was apparent. Nevertheless, elevated IL12B expression in Rgs2−/− animals raises the possibility that RGS2 could dampen Th1 responses. These findings indicate that up-regulation of RGS2, as occurs in response to inhaled corticosteroids and long-acting β2-adrenoceptor agonists, may be beneficial in acute neutrophilic exacerbations of airway disease, including asthma. Electronic supplementary material The online version of this article (10.1186/s13223-018-0266-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tresa George
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mainak Chakraborty
- 2Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mark A Giembycz
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Robert Newton
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| |
Collapse
|
32
|
Rider CF, Altonsy MO, Mostafa MM, Shah SV, Sasse S, Manson ML, Yan D, Kärrman-Mårdh C, Miller-Larsson A, Gerber AN, Giembycz MA, Newton R. Long-Acting β2-Adrenoceptor Agonists Enhance Glucocorticoid Receptor (GR)-Mediated Transcription by Gene-Specific Mechanisms Rather Than Generic Effects via GR. Mol Pharmacol 2018; 94:1031-1046. [PMID: 29959223 PMCID: PMC7385531 DOI: 10.1124/mol.118.112755] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
In asthma, the clinical efficacy of inhaled corticosteroids (ICSs) is enhanced by long-acting β2-adrenoceptor agonists (LABAs). ICSs, or more accurately, glucocorticoids, promote therapeutically relevant changes in gene expression, and, in primary human bronchial epithelial cells (pHBECs) and airway smooth muscle cells, this genomic effect can be enhanced by a LABA. Modeling this interaction in human bronchial airway epithelial BEAS-2B cells transfected with a 2× glucocorticoid response element (2×GRE)-driven luciferase reporter showed glucocorticoid-induced transcription to be enhanced 2- to 3-fold by LABA. This glucocorticoid receptor (GR; NR3C1)-dependent effect occurred rapidly, was insensitive to protein synthesis inhibition, and was maximal when glucocorticoid and LABA were added concurrently. The ability of LABA to enhance GR-mediated transcription was not associated with changes in GR expression, serine (Ser203, Ser211, Ser226) phosphorylation, ligand affinity, or nuclear translocation. Chromatin immunoprecipitation demonstrated that glucocorticoid-induced recruitment of GR to the integrated 2×GRE reporter and multiple gene loci, whose mRNAs were unaffected or enhanced by LABA, was also unchanged by LABA. Transcriptomic analysis revealed glucocorticoid-induced mRNAs were variably enhanced, unaffected, or repressed by LABA. Thus, events leading to GR binding at target genes are not the primary explanation for how LABAs modulate GR-mediated transcription. As many glucocorticoid-induced genes are independently induced by LABA, gene-specific control by GR- and LABA-activated transcription factors may explain these observations. Because LABAs promote similar effects in pHBECs, therapeutic relevance is likely. These data illustrate the need to understand gene function(s), and the mechanisms leading to gene-specific induction, if existing ICS/LABA combination therapies are to be improved.
Collapse
Affiliation(s)
- Christopher F Rider
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Mohammed O Altonsy
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Mahmoud M Mostafa
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Suharsh V Shah
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Sarah Sasse
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Martijn L Manson
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Dong Yan
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Carina Kärrman-Mårdh
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Anna Miller-Larsson
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Anthony N Gerber
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Mark A Giembycz
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| | - Robert Newton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada (C.F.R., M.O.A., M.M.M., S.V.S., D.Y., M.A.G., R.N.); Department of Zoology, Sohag University, Sohag, Egypt (M.O.A.); Department of Medicine, National Jewish Health, Denver, Colorado (S.S., A.N.G.); and Bioscience, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit (M.L.M., C.K.-M.), and Respiratory GMed (A.M.-L.), AstraZeneca, Gothenburg, Molndal, Sweden
| |
Collapse
|
33
|
Westbury GLM, Blais CM, Davis BE, Cockcroft DW. Bronchoprotective effect of vilanterol against methacholine-induced bronchoconstriction in mild asthmatics: A randomized three-way crossover study. Ann Allergy Asthma Immunol 2018; 121:328-332. [PMID: 30017826 DOI: 10.1016/j.anai.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/07/2018] [Accepted: 04/07/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Ultra-long-acting β2 agonists (uLABA) are relatively new anti-asthma medications of which there are three different formulations currently available: olodaterol, indacaterol, and vilanterol. The first 2 formulations have been shown to exert bronchoprotective effects; they are able to prevent airway smooth muscle contraction on exposure to constricting stimuli. However, studies have found that these 2 drugs produce different degrees and durations of bronchoprotection against methacholine. OBJECTIVE The objective of this study was to investigate the degree of bronchoprotection provided by vilanterol against methacholine-induced bronchoconstriction. METHODS Fourteen patients with mild-to-moderate asthma (8 male; baseline percent predicted forced expiratory volume in 1 second [FEV1] > 65%; provocative concentration of methacholine causing a 20% reduction in FEV1 [PC20] ≤ 8 mg/mL) completed this randomized, double-blind, 3-way crossover study. Methacholine challenges were performed before treatment administration (placebo, 100 μg fluticasone furoate, or 25 μg vilanterol + 100 μg fluticasone furoate) and at 0.5 and 24 hours posttreatment. Each treatment arm was separated by a minimum 7-day washout period. A combination therapy of vilanterol+fluticasone furoate was used, because vilanterol is not available as a monotherapy. RESULTS Significant bronchoprotection was evident after the combination treatment at both 0.5 and 24 hours with doubling dose shifts in methacholine PC20 of 2.0 (P = .0004) and 1.6 (P = .0001), respectively. Clinically significant bronchodilation was only recorded at 24 hours after combination treatment (P < .05). CONCLUSION These findings suggest that vilanterol (in combination with fluticasone furoate) provides significant bronchoprotection against methacholine-induced bronchoconstriction for at least 24 hours in patients with mild-to-moderate asthma. CLINICAL TRIAL REGISTRATION clinicaltrials.gov (NCT03315000).
Collapse
Affiliation(s)
- Grace L M Westbury
- Department of Physiology, Division of Respirology, Critical Care and Sleep Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | - Beth E Davis
- Department of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Donald W Cockcroft
- Department of Physiology, Division of Respirology, Critical Care and Sleep Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada; Department of Medicine, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
34
|
Yan D, Hamed O, Joshi T, Mostafa MM, Jamieson KC, Joshi R, Newton R, Giembycz MA. Analysis of the Indacaterol-Regulated Transcriptome in Human Airway Epithelial Cells Implicates Gene Expression Changes in the Adverse and Therapeutic Effects of β2-Adrenoceptor Agonists. J Pharmacol Exp Ther 2018; 366:220-236. [PMID: 29653961 DOI: 10.1124/jpet.118.249292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
The contribution of gene expression changes to the adverse and therapeutic effects of β2-adrenoceptor agonists in asthma was investigated using human airway epithelial cells as a therapeutically relevant target. Operational model-fitting established that the long-acting β2-adrenoceptor agonists (LABA) indacaterol, salmeterol, formoterol, and picumeterol were full agonists on BEAS-2B cells transfected with a cAMP-response element reporter but differed in efficacy (indacaterol ≥ formoterol > salmeterol ≥ picumeterol). The transcriptomic signature of indacaterol in BEAS-2B cells identified 180, 368, 252, and 10 genes that were differentially expressed (>1.5- to <0.67-fold) after 1-, 2-, 6-, and 18-hour of exposure, respectively. Many upregulated genes (e.g., AREG, BDNF, CCL20, CXCL2, EDN1, IL6, IL15, IL20) encode proteins with proinflammatory activity and are annotated by several, enriched gene ontology (GO) terms, including cellular response to interleukin-1, cytokine activity, and positive regulation of neutrophil chemotaxis The general enriched GO term extracellular space was also associated with indacaterol-induced genes, and many of those, including CRISPLD2, DMBT1, GAS1, and SOCS3, have putative anti-inflammatory, antibacterial, and/or antiviral activity. Numerous indacaterol-regulated genes were also induced or repressed in BEAS-2B cells and human primary bronchial epithelial cells by the low efficacy LABA salmeterol, indicating that this genomic effect was neither unique to indacaterol nor restricted to the BEAS-2B airway epithelial cell line. Collectively, these data suggest that the consequences of inhaling a β2-adrenoceptor agonist may be complex and involve widespread changes in gene expression. We propose that this genomic effect represents a generally unappreciated mechanism that may contribute to the adverse and therapeutic actions of β2-adrenoceptor agonists in asthma.
Collapse
Affiliation(s)
- Dong Yan
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Omar Hamed
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Taruna Joshi
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mahmoud M Mostafa
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kyla C Jamieson
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Radhika Joshi
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Calzetta L, Matera MG, Facciolo F, Cazzola M, Rogliani P. Beclomethasone dipropionate and formoterol fumarate synergistically interact in hyperresponsive medium bronchi and small airways. Respir Res 2018; 19:65. [PMID: 29650006 PMCID: PMC5897944 DOI: 10.1186/s12931-018-0770-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/02/2018] [Indexed: 12/15/2022] Open
Abstract
Background Corticosteroids increase the expression of β2-adrenoceptors (β2-ARs) and protect them against down-regulation. Conversely, β2-AR agonists improve the anti-inflammatory action of corticosteroids. Nevertheless, it is still uncertain whether adding a long-acting β2-AR agonist (LABA) to an inhaled corticosteroid (ICS) results in an additive effect, or there is true synergy. Therefore, the aim of this study was to pharmacologically characterize the interaction between the ICS beclomethasone diproprionate (BDP) and the LABA formoterol fumarate (FF) in a validated human ex vivo model of bronchial asthma. Methods Human medium and small airways were stimulated by histamine and treated with different concentrations of BDP and FF, administered alone and in combination at concentration-ratio reproducing ex vivo that of the currently available fixed-dose combination (FDC; BDP/FF 100:6 combination-ratio). Experiments were performed in non-sensitized (NS) and passively sensitized (PS) airways. The pharmacological interaction was assessed by using Bliss Independence and Unified Theory equations. Results BDP/FF synergistically increased the overall bronchorelaxation in NS and PS airways (+ 15.15% ± 4.02%; P < 0.05 vs. additive effect). At low-to-medium concentrations the synergistic interaction was greater in PS than in NS bronchioles (+ 16.68% ± 3.02% and + 7.27% ± 3.05%, respectively). In PS small airways a very strong synergistic interaction (Combination Index: 0.08; + 20.04% ± 2.18% vs. additive effect) was detected for the total concentrations of BDP/FF combination corresponding to 10.6 ng/ml. Conclusion BDP/FF combination synergistically relaxed human bronchi; the extent of such an interaction was very strong at low-to-medium concentrations in PS small airways. Trial registration Not applicable. Electronic supplementary material The online version of this article (10.1186/s12931-018-0770-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luigino Calzetta
- Unit of Respiratory Medicine, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesco Facciolo
- Thoracic Surgery Unit, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
36
|
Newton R. Regulators of G-Protein Signaling as Asthma Therapy? Am J Respir Cell Mol Biol 2018; 58:7-9. [PMID: 29286861 DOI: 10.1165/rcmb.2017-0333ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Robert Newton
- 1 Cumming School of Medicine University of Calgary Calgary, Alberta, Canada
| |
Collapse
|
37
|
Madigan LA, Wong GS, Gordon EM, Chen WS, Balenga N, Koziol-White CJ, Panettieri RA, Levine SJ, Druey KM. RGS4 Overexpression in Lung Attenuates Airway Hyperresponsiveness in Mice. Am J Respir Cell Mol Biol 2018; 58:89-98. [PMID: 28853915 DOI: 10.1165/rcmb.2017-0109oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A cardinal feature of asthma is airway hyperresponsiveness (AHR) to spasmogens, many of which activate G protein-coupled receptors (GPCRs) on airway smooth muscle (ASM) cells. Asthma subtypes associated with allergy are characterized by eosinophilic inflammation in the lung due to the type 2 immune response to allergens and proinflammatory mediators that promote AHR. The degree to which intrinsic abnormalities of ASM contribute to this phenotype remains unknown. The regulators of G protein signaling (RGS) proteins are a large group of intracellular proteins that inhibit GPCR signaling pathways. RGS2- and RGS5-deficient mice develop AHR spontaneously. Although RGS4 is upregulated in ASM from patients with severe asthma, the effects of increased RGS4 expression on AHR in vivo are unknown. Here, we examined the impact of forced RGS4 overexpression in lung on AHR using transgenic (Tg) mice. Tg RGS4 was expressed in bronchial epithelium and ASM in vivo, and protein expression in lung was increased at least 4-fold in Tg mice compared with wild-type (WT) mice. Lung slices from Tg mice contracted less in response to the m3 muscarinic receptor agonist methacholine compared with the WT, although airway resistance in live, unchallenged mice of both strains was similar. Tg mice were partially protected against AHR induced by fungal allergen challenge due to weakened contraction signaling in ASM and reduced type 2 cytokine (IL-5 and IL-13) levels in Tg mice compared with the WT. These results provide support for the hypothesis that increasing RGS4 expression and/or function could be a viable therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Laura A Madigan
- 1 Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Gordon S Wong
- 1 Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Elizabeth M Gordon
- 2 Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Wei-Sheng Chen
- 1 Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Nariman Balenga
- 1 Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Cynthia J Koziol-White
- 3 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Reynold A Panettieri
- 3 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Stewart J Levine
- 2 Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Kirk M Druey
- 1 Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, and
| |
Collapse
|
38
|
Huff RD, Rider CF, Yan D, Newton R, Giembycz MA, Carlsten C, Hirota JA. Inhibition of ABCC4 potentiates combination beta agonist and glucocorticoid responses in human airway epithelial cells. J Allergy Clin Immunol 2017; 141:1127-1130.e5. [PMID: 29103996 DOI: 10.1016/j.jaci.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 09/27/2017] [Accepted: 10/04/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Ryan D Huff
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher F Rider
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dong Yan
- Department of Physiology & Pharmacology, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Department of Cell Biology & Anatomy, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Department of Physiology & Pharmacology, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Chris Carlsten
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeremy A Hirota
- Firestone Institute for Respiratory Health, Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
39
|
Abstract
β2-adrenoceptor agonists, often used in combination with corticosteroids, have been extensively used for the treatment of asthma. However, concerns have been raised regarding their adverse effects and safety including poor asthma control, life-threatening exacerbations, exacerbations that often require hospitalization, and asthma-related deaths. The question as to whether these adverse effects relate to the loss of their bronchoprotective action remains an interesting possibility. In the chapter, we will review the experimental evidence that describes the different potential factors and associated mechanisms that can blunt the therapeutic action of β2-adrenoceptor agonists in asthma. We show here evidence that various key inflammatory cytokines, growth factors, some respiratory viruses, certain allergens, unknown factors present in serum from atopic asthmatics have the capacity to impair β2-adrenoceptor function in airway smooth muscle, the main target of these drugs. More importantly, we present our latest research describing the role played by mast cells in impairing β2-adrenoceptor function. Although no definitive conclusion could be made regarding the implication of one single mechanism, receptor uncoupling, or receptor desensitization due to phosphorylation represents the main inhibitory pathways associated with a loss of β2-adrenoceptor function in airway smooth muscle. Targeting the pathways leading to β2-adrenoceptor dysfunction will likely provide novel therapies to improve the efficacy of β2-agonists in asthma.
Collapse
|
40
|
Shaikh N, Johnson M, Hall DA, Chung KF, Riley JH, Worsley S, Bhavsar PK. Intracellular interactions of umeclidinium and vilanterol in human airway smooth muscle. Int J Chron Obstruct Pulmon Dis 2017; 12:1903-1913. [PMID: 28721035 PMCID: PMC5501633 DOI: 10.2147/copd.s134420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Intracellular mechanisms of action of umeclidinium (UMEC), a long-acting muscarinic receptor antagonist, and vilanterol (VI), a long-acting β2-adrenoceptor (β2R) agonist, were investigated in target cells: human airway smooth-muscle cells (ASMCs). Materials and methods ASMCs from tracheas of healthy lung-transplant donors were treated with VI, UMEC, UMEC and VI combined, or control compounds (salmeterol, propranolol, ICI 118.551, or methacholine [MCh]). Cyclic adenosine monophosphate (cAMP) was measured using an enzyme-linked immunosorbent assay, intracellular free calcium ([Ca2+]i) using a fluorescence assay, and regulator of G-protein signaling 2 (RGS2) messenger RNA using real-time quantitative polymerase chain reaction. Results VI and salmeterol (10−12–10−6 M) induced cAMP production from ASMCs in a concentration-dependent manner, which was greater for VI at all concentrations. β2R antagonism by propranolol or ICI 118.551 (10−12–10−4 M) resulted in concentration-dependent inhibition of VI-induced cAMP production, and ICI 118.551 was more potent. MCh (5×10−6 M, 30 minutes) attenuated VI-induced cAMP production (P<0.05), whereas pretreatment with UMEC (10−8 M, 1 hour) restored the magnitude of VI-induced cAMP production. ASMC stimulation with MCh (10−11–5×10−6 M) resulted in a concentration-dependent increase in [Ca2+]i, which was attenuated with UMEC pretreatment. Reduction of MCh-induced [Ca2+]i release was greater with UMEC + VI versus UMEC. UMEC enhanced VI-induced RGS2 messenger RNA expression. Conclusion These data indicate that UMEC reverses cholinergic inhibition of VI-induced cAMP production, and is a more potent muscarinic receptor antagonist when in combination with VI versus either alone.
Collapse
Affiliation(s)
- Nooreen Shaikh
- Experimental Studies, National Heart and Lung Institute, Imperial College London.,Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London
| | | | - David A Hall
- Fibrosis and Lung Injury Development Planning Unit, GlaxoSmithKline, Stevenage
| | - Kian Fan Chung
- Experimental Studies, National Heart and Lung Institute, Imperial College London.,Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London
| | - John H Riley
- Respiratory Global Franchise, GlaxoSmithKline, Uxbridge
| | - Sally Worsley
- Respiratory Research & Development, GlaxoSmithKline, Uxbridge, UK
| | - Pankaj K Bhavsar
- Experimental Studies, National Heart and Lung Institute, Imperial College London.,Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London
| |
Collapse
|
41
|
Kua KP, Lee SWH. Systematic Review and Meta-Analysis of the Efficacy and Safety of Combined Epinephrine and Corticosteroid Therapy for Acute Bronchiolitis in Infants. Front Pharmacol 2017; 8:396. [PMID: 28690542 PMCID: PMC5479924 DOI: 10.3389/fphar.2017.00396] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/06/2017] [Indexed: 01/28/2023] Open
Abstract
Objective: To evaluate the effectiveness of combined epinephrine and corticosteroid therapy for acute bronchiolitis in infants. Methods: Four electronic databases (MEDLINE, EMBASE, CINAHL, and CENTRAL) were searched from their inception to February 28, 2017 for studies involving infants aged less than 24 months with bronchiolitis which assessed the use of epinephrine and corticosteroid combination therapy. The methodological quality of the included studies was assessed using the Cochrane Collaboration's Risk of Bias Tool. A random-effects meta-analysis was used to pool the effect estimates. The primary outcomes were hospital admission rate and length of hospital stay. Results: Of 1,489 citations identified, 5 randomized controlled trials involving 1,157 patients were included. All studies were of high quality and low risk of bias. Results of the meta-analysis showed no significant differences in the primary outcomes. Hospitalization rate was reduced by combinatorial therapy of epinephrine and corticosteroid in only one out of five studies, whereas pooled data indicated no benefit over epinephrine plus placebo. Clinical severity scores were significantly improved in all five RCTs when assessed individually, but no benefit was observed compared to epinephrine monotherapy when the data were pooled together. Pooled data showed that combination therapy was more effective at improving oxygen saturation level (mean difference: −0.70; 95% confidence interval: −1.17 to −0.22, p = 0.004). There was no difference in the risk of serious adverse events in infants treated with the combined epinephrine and corticosteroid therapy. Conclusions: Combination treatment of epinephrine and dexamethasone was ineffective in reducing hospital admission and length of stay among infants with bronchiolitis.
Collapse
Affiliation(s)
- Kok P Kua
- School of Pharmacy, Monash University MalaysiaBandar Sunway, Malaysia.,Department of Pharmacy, Petaling District Health Office, Ministry of Health MalaysiaPetaling Jaya, Malaysia
| | - Shaun W H Lee
- School of Pharmacy, Monash University MalaysiaBandar Sunway, Malaysia
| |
Collapse
|
42
|
Keränen T, Hömmö T, Moilanen E, Korhonen R. β 2-receptor agonists salbutamol and terbutaline attenuated cytokine production by suppressing ERK pathway through cAMP in macrophages. Cytokine 2017; 94:1-7. [PMID: 28162907 DOI: 10.1016/j.cyto.2016.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/17/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023]
Abstract
β2-receptor agonists are used in the treatment of inflammatory obstructive lung diseases asthma and COPD as a symptomatic remedy, but they have been suggested to possess anti-inflammatory properties, also. β2-receptor activation is considered to lead to the activation of ERK pathway through G-protein- and cAMP-independent mechanisms. In this study, we investigated the effects of β2-receptor agonists salbutamol and terbutaline on the production of inflammatory factors in macrophages. We found that β2-receptor agonists inhibited LPS-induced ERK phosphorylation and the production of MCP-1. A chemical cAMP analog 8-Br-cAMP also inhibited ERK phosphorylation and TNF and MCP-1 release. As expected, MAPK/ERK kinase (MEK)1/2 inhibitor PD0325901 inhibited ERK phosphorylation and suppressed both TNF and MCP-1 production. In conclusion, we suggest that β2-receptor agonists salbutamol and terbutaline inhibit inflammatory gene expression partly by a mechanism dependent on cAMP leading to the inhibition of ERK signaling in macrophages. Observed anti-inflammatory effects of β2-receptor agonists may contribute to the clinical effects of these drugs.
Collapse
Affiliation(s)
- Tiina Keränen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, Tampere, Finland
| | - Tuija Hömmö
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, Tampere, Finland
| | - Riku Korhonen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
43
|
Joshi T, Yan D, Hamed O, Tannheimer SL, Phillips GB, Wright CD, Kim M, Salmon M, Newton R, Giembycz MA. GS-5759, a Bifunctional β2-Adrenoceptor Agonist and Phosphodiesterase 4 Inhibitor for Chronic Obstructive Pulmonary Disease with a Unique Mode of Action: Effects on Gene Expression in Human Airway Epithelial Cells. J Pharmacol Exp Ther 2017; 360:324-340. [PMID: 27927912 DOI: 10.1124/jpet.116.237743] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/05/2016] [Indexed: 03/08/2025] Open
Abstract
(R)-6-[(3-{[4-(5-{[2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethyl]amino}pent-1-yn-1-yl)phenyl] carbamoyl}phenyl)sulphonyl]-4-[(3-methoxyphenyl)amino]-8-methylquinoline-3-carboxamide trifluoroacetic acid (GS-5759) is a bifunctional ligand composed of a quinolinone-containing pharmacophore [β2-adrenoceptor agonist orthostere (β2A)] found in several β2-adrenoceptor agonists, including indacaterol, linked covalently to a phosphodiesterase 4 (PDE4) inhibitor related to 6-[3-(dimethylcarbamoyl)benzenesulphonyl]-4-[(3-methoxyphenyl)amino]-8-methylquinoline-3-carboxamide (GSK 256066) by a pent-1-yn-1-ylbenzene spacer. GS-5759 had a similar affinity for PDE4B1 and the native β2-adrenoceptor expressed on BEAS-2B human airway epithelial cells. However, compared with the monofunctional parent compound, β2A, the KA of GS-5759 for the β2-adrenoceptor was 35-fold lower. Schild analysis determined that the affinities of the β-adrenoceptor antagonists, (2R,3R)-1-[(2,3-dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl) amino]-2-butanol (ICI 118551) and propranolol, were agonist-dependent, being significantly lower for GS-5759 than β2A. Collectively, these data can be explained by "forced proximity," bivalent binding where the pharmacophore in GS-5759 responsible for PDE4 inhibition also interacts with a nonallosteric domain within the β2-adrenoceptor that enhances the affinity of β2A for the orthosteric site. Microarray analyses revealed that, after 2-hour exposure, GS-5759 increased the expression of >3500 genes in BEAS-2B cells that were highly rank-order correlated with gene expression changes produced by indacaterol and GSK 256066 in combination (Ind/GSK). Moreover, the line of regression began close to the origin with a slope of 0.88, indicating that the magnitude of most gene expression changes produced by Ind/GSK was quantitatively replicated by GS-5759. Thus, GS-5759 is a novel compound exhibiting dual β2-adrenoceptor agonism and PDE4 inhibition with potential to interact on target tissues in a synergistic manner. Such polypharmacological behavior may be particularly effective in chronic obstructive pulmonary disease and other complex disorders where multiple processes interact to promote disease pathogenesis and progression.
Collapse
Affiliation(s)
- Taruna Joshi
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Dong Yan
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Omar Hamed
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Stacey L Tannheimer
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Gary B Phillips
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Clifford D Wright
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Musong Kim
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Michael Salmon
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Robert Newton
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| | - Mark A Giembycz
- Departments of Physiology and Pharmacology (T.J., D.Y., O.H., M.A.G.) and Cell Biology and Anatomy (R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Translational Medicine, Biomarkers (S.L.T.), Inflammation Research (C.D.W., M.S.), and Medicinal Chemistry (G.B.P., M.K.), Gilead Sciences Inc., Seattle, Washington
| |
Collapse
|
44
|
George T, Bell M, Chakraborty M, Siderovski DP, Giembycz MA, Newton R. Protective Roles for RGS2 in a Mouse Model of House Dust Mite-Induced Airway Inflammation. PLoS One 2017; 12:e0170269. [PMID: 28107494 PMCID: PMC5249169 DOI: 10.1371/journal.pone.0170269] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/30/2016] [Indexed: 12/16/2022] Open
Abstract
The GTPase-accelerating protein, regulator of G-protein signalling 2 (RGS2) reduces signalling from G-protein-coupled receptors (GPCRs) that signal via Gαq. In humans, RGS2 expression is up-regulated by inhaled corticosteroids (ICSs) and long-acting β2-adrenoceptor agonists (LABAs) such that synergy is produced in combination. This may contribute to the superior clinical efficacy of ICS/LABA therapy in asthma relative to ICS alone. In a murine model of house dust mite (HDM)-induced airways inflammation, three weeks of intranasal HDM (25 μg, 3×/week) reduced lung function and induced granulocytic airways inflammation. Compared to wild type animals, Rgs2-/- mice showed airways hyperresponsiveness (increased airways resistance and reduced compliance). While HDM increased pulmonary inflammation observed on hematoxylin and eosin-stained sections, there was no difference between wild type and Rgs2-/- animals. HDM-induced mucus hypersecretion was also unaffected by RGS2 deficiency. However, inflammatory cell counts in the bronchoalveolar lavage fluid of Rgs2-/- animals were significantly increased (57%) compared to wild type animals and this correlated with increased granulocyte (neutrophil and eosinophil) numbers. Likewise, cytokine and chemokine (IL4, IL17, IL5, LIF, IL6, CSF3, CXCLl, CXCL10 and CXCL11) release was increased by HDM exposure. Compared to wild type, Rgs2-/- animals showed a trend towards increased expression for many cytokines/chemokines, with CCL3, CCL11, CXCL9 and CXCL10 being significantly enhanced. As RGS2 expression was unaffected by HDM exposure, these data indicate that RGS2 exerts tonic bronchoprotection in HDM-induced airways inflammation. Modest anti-inflammatory and anti-remodelling roles for RGS2 are also suggested. If translatable to humans, therapies that maximize RGS2 expression may prove advantageous.
Collapse
Affiliation(s)
- Tresa George
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Matthew Bell
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Mainak Chakraborty
- Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - David P. Siderovski
- Blanchette Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Mark A. Giembycz
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
45
|
Abstract
Viral bronchiolitis is a common clinical syndrome affecting infants and young children. Concern about its associated morbidity and cost has led to a large body of research that has been summarised in systematic reviews and integrated into clinical practice guidelines in several countries. The evidence and guideline recommendations consistently support a clinical diagnosis with the limited role for diagnostic testing for children presenting with the typical clinical syndrome of viral upper respiratory infection progressing to the lower respiratory tract. Management is largely supportive, focusing on maintaining oxygenation and hydration of the patient. Evidence suggests no benefit from bronchodilator or corticosteroid use in infants with a first episode of bronchiolitis. Evidence for other treatments such as hypertonic saline is evolving but not clearly defined yet. For infants with severe disease, the insufficient available data suggest a role for high-flow nasal cannula and continuous positive airway pressure use in a monitored setting to prevent respiratory failure.
Collapse
Affiliation(s)
- Todd A Florin
- Division of Pediatric Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Amy C Plint
- Division of Emergency Medicine, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada; Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Joseph J Zorc
- Division of Emergency Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
46
|
Altonsy MO, Mostafa MM, Gerber AN, Newton R. Long-acting β 2-agonists promote glucocorticoid-mediated repression of NF-κB by enhancing expression of the feedback regulator TNFAIP3. Am J Physiol Lung Cell Mol Physiol 2016; 312:L358-L370. [PMID: 28039105 DOI: 10.1152/ajplung.00426.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Glucocorticoids, or corticosteroids, are effective treatments for many chronic inflammatory diseases, and in mild/moderate asthma, long-acting β2-adrenoceptor agonists (LABAs) enhance the efficacy of inhaled corticosteroids (ICSs) more than increasing the ICS dose. In human bronchial epithelial, BEAS-2B, cells, expression of TNFα-induced protein-3 (TNFAIP3), or A20, a dual-ubiquitin ligase that provides feedback inhibition of NF-κB, was induced by budesonide, an ICS, and formoterol, a LABA, and was further enhanced by budesonide-formoterol combination. The proinflammatory cytokine TNF induced TNFAIP3 and TNF expression. Whereas subsequent budesonide treatment enhanced TNF-induced TNFAIP3 and reduced TNF expression, formoterol amplified these differential effects. In primary human airway smooth muscle cells, TNFAIP3 expression was induced by TNF. This was largely unaffected by budesonide but was acutely enhanced by budesonide-formoterol combination. In BEAS-2B cells, TNF recruited RELA, the main NF-κB transactivating subunit, to a 3' region of the TNF gene. RELA binding was reduced by budesonide, was further reduced by formoterol cotreatment, and was associated with reduced RNA polymerase II recruitment to the TNF gene. This is consistent with reduced TNF expression. TNFAIP3 knockdown enhanced TNF expression in the presence of TNF, TNF plus budesonide, and TNF plus budesonide-formoterol combination and confirms feedback inhibition. A luciferase reporter containing the TNF 3' RELA binding region recapitulated TNF inducibility and was inhibited by an IκB kinase inhibitor and TNFAIP3 overexpression. Repression of reporter activity by budesonide was increased by formoterol and involved TNFAIP3. Thus LABAs may improve the anti-inflammatory properties of ICSs by augmenting TNFAIP3 expression to negatively regulate NF-κB.
Collapse
Affiliation(s)
- Mohammed O Altonsy
- Department of Cell Biology and Anatomy, Airway Inflammation Research Group, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Zoology, Sohag University, Sohag, Egypt
| | - Mahmoud M Mostafa
- Department of Cell Biology and Anatomy, Airway Inflammation Research Group, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anthony N Gerber
- Department of Medicine, National Jewish Health, Denver, Colorado; and.,Department of Medicine, University of Colorado, Denver, Colorado
| | - Robert Newton
- Department of Cell Biology and Anatomy, Airway Inflammation Research Group, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada;
| |
Collapse
|
47
|
Newton R, Giembycz MA. Understanding how long-acting β 2 -adrenoceptor agonists enhance the clinical efficacy of inhaled corticosteroids in asthma - an update. Br J Pharmacol 2016; 173:3405-3430. [PMID: 27646470 DOI: 10.1111/bph.13628] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/19/2016] [Accepted: 08/21/2016] [Indexed: 12/18/2022] Open
Abstract
In moderate-to-severe asthma, adding an inhaled long-acting β2 -adenoceptor agonist (LABA) to an inhaled corticosteroid (ICS) provides better disease control than simply increasing the dose of ICS. Acting on the glucocorticoid receptor (GR, gene NR3C1), ICSs promote anti-inflammatory/anti-asthma gene expression. In vitro, LABAs synergistically enhance the maximal expression of many glucocorticoid-induced genes. Other genes, including dual-specificity phosphatase 1(DUSP1) in human airways smooth muscle (ASM) and epithelial cells, are up-regulated additively by both drug classes. Synergy may also occur for LABA-induced genes, as illustrated by the bronchoprotective gene, regulator of G-protein signalling 2 (RGS2) in ASM. Such effects cannot be produced by either drug alone and may explain the therapeutic efficacy of ICS/LABA combination therapies. While the molecular basis of synergy remains unclear, mechanistic interpretations must accommodate gene-specific regulation. We explore the concept that each glucocorticoid-induced gene is an independent signal transducer optimally activated by a specific, ligand-directed, GR conformation. In addition to explaining partial agonism, this realization provides opportunities to identify novel GR ligands that exhibit gene expression bias. Translating this into improved therapeutic ratios requires consideration of GR density in target tissues and further understanding of gene function. Similarly, the ability of a LABA to interact with a glucocorticoid may be suboptimal due to low β2 -adrenoceptor density or biased β2 -adrenoceptor signalling. Strategies to overcome these limitations include adding-on a phosphodiesterase inhibitor and using agonists of other Gs-coupled receptors. In all cases, the rational design of ICS/LABA, and derivative, combination therapies requires functional knowledge of induced (and repressed) genes for therapeutic benefit to be maximized.
Collapse
Affiliation(s)
- Robert Newton
- Department of Cell Biology and Anatomy, Airways Inflammation Research Group, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
48
|
Pera T, Penn RB. Bronchoprotection and bronchorelaxation in asthma: New targets, and new ways to target the old ones. Pharmacol Ther 2016; 164:82-96. [PMID: 27113408 PMCID: PMC4942340 DOI: 10.1016/j.pharmthera.2016.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023]
Abstract
Despite over 50years of inhaled beta-agonists and corticosteroids as the default management or rescue drugs for asthma, recent research suggests that new therapeutic options are likely to emerge. This belief stems from both an improved understanding of what causes and regulates airway smooth muscle (ASM) contraction, and the identification of new targets whose inhibition or activation can relax ASM. In this review we discuss the recent findings that provide new insight into ASM contractile regulation, a revolution in pharmacology that identifies new ways to "tune" G protein-coupled receptors to improve therapeutic efficacy, and the discovery of several novel targets/approaches capable of effecting bronchoprotection or bronchodilation.
Collapse
Affiliation(s)
- Tonio Pera
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Raymond B Penn
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
49
|
Leigh R, Mostafa MM, King EM, Rider CF, Shah S, Dumonceaux C, Traves SL, McWhae A, Kolisnik T, Kooi C, Slater DM, Kelly MM, Bieda M, Miller-Larsson A, Newton R. An inhaled dose of budesonide induces genes involved in transcription and signaling in the human airways: enhancement of anti- and proinflammatory effector genes. Pharmacol Res Perspect 2016; 4:e00243. [PMID: 28116096 PMCID: PMC5242176 DOI: 10.1002/prp2.243] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/03/2016] [Indexed: 12/27/2022] Open
Abstract
Although inhaled glucocorticoids, or corticosteroids (ICS), are generally effective in asthma, understanding their anti‐inflammatory actions in vivo remains incomplete. To characterize glucocorticoid‐induced modulation of gene expression in the human airways, we performed a randomized placebo‐controlled crossover study in healthy male volunteers. Six hours after placebo or budesonide inhalation, whole blood, bronchial brushings, and endobronchial biopsies were collected. Microarray analysis of biopsy RNA, using stringent (≥2‐fold, 5% false discovery rate) or less stringent (≥1.25‐fold, P ≤ 0.05) criteria, identified 46 and 588 budesonide‐induced genes, respectively. Approximately two third of these genes are transcriptional regulators (KLF9, PER1, TSC22D3, ZBTB16), receptors (CD163, CNR1, CXCR4, LIFR, TLR2), or signaling genes (DUSP1, NFKBIA, RGS1, RGS2, ZFP36). Listed genes were qPCR verified. Expression of anti‐inflammatory and other potentially beneficial genes is therefore confirmed and consistent with gene ontology (GO) terms for negative regulation of transcription and gene expression. However, GO terms for transcription, signaling, metabolism, proliferation, inflammatory responses, and cell movement were also associated with the budesonide‐induced genes. The most enriched functional cluster indicates positive regulation of proliferation, locomotion, movement, and migration. Moreover, comparison with the budesonide‐induced expression profile in primary human airway epithelial cells shows considerable cell type specificity. In conclusion, increased expression of multiple genes, including the transcriptional repressor, ZBTB16, that reduce inflammatory signaling and gene expression, occurs in the airways and blood and may contribute to the therapeutic efficacy of ICS. This provides a previously lacking insight into the in vivo effects of ICS and should promote strategies to improve glucocorticoid efficacy in inflammatory diseases.
Collapse
Affiliation(s)
- Richard Leigh
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Mahmoud M Mostafa
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Elizabeth M King
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Christopher F Rider
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Suharsh Shah
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Curtis Dumonceaux
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Suzanne L Traves
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Andrew McWhae
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Tyler Kolisnik
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | - Cora Kooi
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Donna M Slater
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | - Margaret M Kelly
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Mark Bieda
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | | | - Robert Newton
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| |
Collapse
|
50
|
Sasse SK, Altonsy MO, Kadiyala V, Cao G, Panettieri RA, Gerber AN. Glucocorticoid and TNF signaling converge at A20 (TNFAIP3) to repress airway smooth muscle cytokine expression. Am J Physiol Lung Cell Mol Physiol 2016; 311:L421-32. [PMID: 27371733 DOI: 10.1152/ajplung.00179.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022] Open
Abstract
Airway smooth muscle is a major target tissue for glucocorticoid (GC)-based asthma therapies, however, molecular mechanisms through which the GC receptor (GR) exerts therapeutic effects in this key airway cell type have not been fully elucidated. We previously identified the nuclear factor-κB (NF-κB) inhibitor, A20 (TNFAIP3), as a mediator of cytokine repression by glucocorticoids (GCs) in airway epithelial cells and defined cooperative regulation of anti-inflammatory genes by GR and NF-κB as a key mechanistic underpinning of airway epithelial GR function. Here, we expand on these findings to determine whether a similar mechanism is operational in human airway smooth muscle (HASM). Using HASM cells derived from normal and fatal asthma samples as an in vitro model, we demonstrate that GCs spare or augment TNF-mediated induction of A20 (TNFAIP3), TNIP1, and NFKBIA, all implicated in negative feedback control of NF-κB-driven inflammatory processes. We applied chromatin immunoprecipitation and reporter analysis to show that GR and NF-κB directly regulate A20 expression in HASM through cooperative induction of an intronic enhancer. Using overexpression, we show for the first time that A20 and its interacting partner, TNIP1, repress TNF signaling in HASM cells. Moreover, we applied small interfering RNA-based gene knockdown to demonstrate that A20 is required for maximal cytokine repression by GCs in HASM. Taken together, our data suggest that inductive regulation of A20 by GR and NF-κB contributes to cytokine repression in HASM.
Collapse
Affiliation(s)
- Sarah K Sasse
- Department of Medicine, National Jewish Health, Denver, Colorado
| | | | - Vineela Kadiyala
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine & Science, Rutgers University, New Brunswick, New Jersey; and
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine & Science, Rutgers University, New Brunswick, New Jersey; and
| | - Anthony N Gerber
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Medicine, University of Colorado, Denver, Colorado
| |
Collapse
|