1
|
Abdelmageed M, Güzelgül F, Yalin S, Akkapulu M. Determination of oxidative stress and copeptin levels of COVID-19 according to the clinical course. Microb Pathog 2024; 199:107263. [PMID: 39736344 DOI: 10.1016/j.micpath.2024.107263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
OBJECTIVE COVID-19, caused by the novel coronavirus SARS-CoV-2, is characterized by hyperinflammation, which can trigger oxidative stress. At the same time, COVID-19 is accompanied by both psychological and physical stress. Copeptin, a novel stress marker, has been shown to predict disease outcomes in stress-induced diseases. In this study, we aimed to explore the potential of copeptin, with inflammatory and oxidative stress markers, in distinguishing between different clinical courses of COVID-19. MATERIALS AND METHODS This case-control study included 75 participants: 25 COVID-19 patients hospitalized in the intensive care unit (ICU), 25 non-ICU COVID-19 patients, and 25 healthy individuals. 64 % of the ICU patients received corticosteroid treatment for 4-10 days before sampling. Serum concentrations of the study parameters were assessed by enzyme-linked immunosorbent assay (ELISA) and compared between the study groups. RESULTS Serum IL-6 levels (p < 0.001) were significantly higher in ICU patients compared to non-ICU patients and the control group. Serum MDA (p < 0.001) and 4-HNE (p = 0.027) concentrations were significantly lower in the ICU group in comparison with the other groups. Copeptin was not statistically significant. MDA (p = 0.040) and 4-HNE (p = 0.017) levels were significantly lower in the treated ICU group than the untreated one. CONCLUSIONS Serum IL-6 levels were noticeably associated with COVID-19 severity. Corticosteroid therapy administration seemed to influence MDA and 4-HNE levels in the treated group, with no obvious influence on IL-6 and copeptin in the same cohort. This data suggests that in SARS-CoV-2 infection, corticosteroids may act through a rapid non-genomic mechanism.
Collapse
Affiliation(s)
- Marwa Abdelmageed
- Department of Medical Biochemistry, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| | - Figen Güzelgül
- Department of Biochemistry, Faculty of Pharmacy, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| | - Serap Yalin
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Türkiye.
| | - Merih Akkapulu
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Türkiye.
| |
Collapse
|
2
|
Srivali N, De Giacomi F, Moua T, Ryu JH. Corticosteroid therapy for treating acute exacerbation of interstitial lung diseases: a systematic review. Thorax 2024:thorax-2024-222636. [PMID: 39721758 DOI: 10.1136/thorax-2024-222636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Acute exacerbation of interstitial lung disease (AE-ILD) often results in death and poses significant challenges in clinical management. While corticosteroids are frequently employed, the optimal regimen and their clinical efficacy remain uncertain. To address this knowledge gap, we undertook a systematic review to evaluate the impact of steroid therapy on clinical outcomes in patients experiencing AE-ILD. METHOD Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, we systematically searched multiple databases, identifying 12 454 articles. After removing duplicates and screening titles and abstracts, 447 articles were selected for full-text review. Ultimately, nine studies met inclusion criteria, comparing high-dose corticosteroids with low-dose or non-steroidal interventions in treating AE-ILD. Key outcomes included in-hospital and long-term mortality, as well as AE recurrence. RESULTS Analysis of nine studies (total n=18 509) revealed differential treatment effects based on the ILD subtype. In non-idiopathic pulmonary fibrosis (IPF) ILD, high-dose corticosteroid therapy (>1.0 mg/kg prednisolone) demonstrated improved survival (adjusted HR 0.221, 95% CI 0.102 to 0.480, p<0.001) and reduced 90-day mortality. Early tapering of high-dose corticosteroids (>10% reduction within 2 weeks) reduced in-hospital mortality (adjusted HR 0.37, 95% CI 0.14 to 0.99). Higher cumulative doses in the first 30 days (5185±2414 mg/month vs 3133±1990 mg/month) were associated with lower recurrence rates (adjusted HR 0.61, 95% CI 0.41 to 0.90, p=0.02). In IPF patients, however, high-dose therapy showed inconsistent benefits, with some studies reporting increased mortality risk (OR 1.075, 95% CI 1.044 to 1.107, p<0.001). CONCLUSION This review emphasises the potential benefits of individualised treatment approaches for AE-ILD but highlights the need for caution in making definitive recommendations. Although high-dose corticosteroids may show promise, particularly in non-IPF cases, the current evidence is inconsistent, and the lack of robust supporting literature makes it difficult to draw firm conclusions. Further research through randomised controlled trials is necessary to refine and optimise therapeutic strategies for AE-ILD.
Collapse
Affiliation(s)
- Narat Srivali
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Teng Moua
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jay H Ryu
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Devillier P, Bardin E, Grassin-Delyle S. Les différents corticoïdes inhalés. Quels avantages respectifs ? REVUE FRANÇAISE D'ALLERGOLOGIE 2023. [DOI: 10.1016/j.reval.2023.103293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
4
|
The Combination of Bioinformatics Analysis and Untargeted Metabolomics Reveals Potential Biomarkers and Key Metabolic Pathways in Asthma. Metabolites 2022; 13:metabo13010025. [PMID: 36676950 PMCID: PMC9860906 DOI: 10.3390/metabo13010025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Asthma is a complex chronic airway inflammatory disease that seriously impacts patients' quality of life. As a novel approach to exploring the pathogenesis of diseases, metabolomics provides the potential to identify biomarkers of asthma host susceptibility and elucidate biological pathways. The aim of this study was to screen potential biomarkers and biological pathways so as to provide possible pharmacological therapeutic targets for asthma. In the present study, we merged the differentially expressed genes (DEGs) of asthma in the GEO database with the metabolic genes obtained by Genecard for bioinformatics analysis and successfully screened out the metabolism-related hub genes (HIF1A, OCRL, NNMT, and PER1). Then, untargeted metabolic techniques were utilized to reveal HDM-induced metabolite alterations in 16HBE cells. A total of 45 significant differential metabolites and 5 differential metabolic pathways between the control group and HDM group were identified based on the OPLS-DA model. Finally, three key metabolic pathways, including glycerophospholipid metabolism, galactose metabolism, and alanine, aspartate, and glutamate metabolism, were screened through the integrated analysis of bioinformatics data and untargeted metabolomics data. Taken together, these findings provide valuable insights into the pathophysiology and targeted therapy of asthma and lay a foundation for further research.
Collapse
|
5
|
Amratia DA, Viola H, Ioachimescu OC. Glucocorticoid therapy in respiratory illness: bench to bedside. J Investig Med 2022; 70:1662-1680. [PMID: 35764344 DOI: 10.1136/jim-2021-002161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2022] [Indexed: 11/07/2022]
Abstract
Each year, hundreds of millions of individuals are affected by respiratory disease leading to approximately 4 million deaths. Most respiratory pathologies involve substantially dysregulated immune processes that either fail to resolve the underlying process or actively exacerbate the disease. Therefore, clinicians have long considered immune-modulating corticosteroids (CSs), particularly glucocorticoids (GCs), as a critical tool for management of a wide spectrum of respiratory conditions. However, the complex interplay between effectiveness, risks and side effects can lead to different results, depending on the disease in consideration. In this comprehensive review, we present a summary of the bench and the bedside evidence regarding GC treatment in a spectrum of respiratory illnesses. We first describe here the experimental evidence of GC effects in the distal airways and/or parenchyma, both in vitro and in disease-specific animal studies, then we evaluate the recent clinical evidence regarding GC treatment in over 20 respiratory pathologies. Overall, CS remain a critical tool in the management of respiratory illness, but their benefits are dependent on the underlying pathology and should be weighed against patient-specific risks.
Collapse
|
6
|
Liu D, Long M, Gao L, Chen Y, Li F, Shi Y, Gu N. Nanomedicines Targeting Respiratory Injuries for Pulmonary Disease Management. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202112258] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 01/02/2025]
Abstract
AbstractThe respiratory system holds crucial importance in the biology of vertebrate animals. Injuries of the respiratory system caused by viral infections (e.g., by COVID‐19, MERS, and SARS) can lead to severe or lethal conditions. So far there are no effective treatments for respiratory injuries. This represents a highly unmet clinical need, e.g., during the current COVID‐19 pandemic. Nanomedicines have high potential in the treatment of respiratory injuries. In this review, the pathology and clinical treatments of major respiratory injuries, acute lung injury, and acute respiratory distress syndrome are briefly summarized. The review primarily focuses on nanomedicines based on liposomes, solid lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, which are tested in preclinical models for the treatment of respiratory injuries. These nanomedicines are utilized to deliver a variety of therapeutic agents, including corticosteroids, statins, and nucleic acids. Furthermore, nanomedicines are also investigated for other respiratory diseases including chronic obstructive pulmonary disease and asthma. The promising preclinical results of various nanoformulations from these studies suggest the potential of nanomedicines for future clinical management of respiratory viral infections and diseases.
Collapse
Affiliation(s)
- Dong Liu
- School of Biological and Pharmaceutical Engineering West Anhui University Lu'An 237012 P. R. China
| | - Mengmeng Long
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biomedical Sciences and Medical Engineering Southeast University Nanjing 210009 P. R. China
| | - Leilei Gao
- School of Biological and Pharmaceutical Engineering West Anhui University Lu'An 237012 P. R. China
| | - Yanjun Chen
- School of Biological and Pharmaceutical Engineering West Anhui University Lu'An 237012 P. R. China
| | - Fang Li
- School of Biological and Pharmaceutical Engineering West Anhui University Lu'An 237012 P. R. China
| | - Yang Shi
- Institute for Experimental Molecular Imaging Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering Faculty of Medicine RWTH Aachen University 52074 Aachen Germany
| | - Ning Gu
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biomedical Sciences and Medical Engineering Southeast University Nanjing 210009 P. R. China
| |
Collapse
|
7
|
Jain S, Durugkar S, Saha P, Gokhale SB, Naidu VGM, Sharma P. Effects of intranasal azithromycin on features of cigarette smoke-induced lung inflammation. Eur J Pharmacol 2022; 915:174467. [PMID: 34478690 DOI: 10.1016/j.ejphar.2021.174467] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/04/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Airflow limitation in chronic obstructive pulmonary disease (COPD) is the result of exaggerated airway fibrosis and obliteration of the small airways due to persistent inflammation, and an impaired anti-oxidant response. EMT has been implicated as an active signalling process in cigarette smoke (CS)-induced lung pathology, and macrolide Azithromycin (AZT) use has gained interest in treating COPD. Here, we tested effectiveness of intra-nasal AZT alone and in combination with dexamethasone (DEX) on CS-induced acute lung inflammation. Human alveolar epithelial cells (A549) were treated with CS extract (CSE) for 48 h, and male Balb/c mice were exposed to CS (3 cigarettes-3 times/day) for 4 days. The effects of AZT alone (0.25 and 1.25 μM, in vitro; 0.5 and 5 mg/kg, in vivo) or in combination with DEX (1 μM, in vitro; 1 mg/kg, in vivo) on CS-induced cellular cytotoxicity, oxidative stress, inflammation, and lung function were assessed. AZT alone and in combination with DEX significantly inhibited the CS (E)-induced expression of mesenchymal protein markers and the regulatory protein β-catenin. Furthermore, AZT by itself or in combination with DEX significantly suppressed CS-induced expression of the proinflammtory cytokines TNFα, IL1β and IL6 and prevented pNFkB. Mechanistically, AZT restored the CS-induced reduction in anti-oxidant transcription factor NRF2 and upregulated HDAC2 levels, thereby repressing inflammatory gene expression. Beneficial effects of AZT functionally translated in improved lung mechanics in vivo. Further preclinical and clinical studies are warranted to fully establish and validate the therapeutic efficacy of AZT as a mono- or combination therapy for the treatment of COPD.
Collapse
Affiliation(s)
- Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Sneha Durugkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Pritam Saha
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Sharad B Gokhale
- Department of Civil Engineering, Indian Institute of Technology Guwahati, North Amingaon, Guwahati, Assam, 781039, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India.
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
8
|
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]
|
9
|
Nitric-Oxide-Releasing Dexamethasone Derivative NCX-1005 Improves Lung Function and Attenuates Inflammation in Experimental Lavage-Induced ARDS. Pharmaceutics 2021; 13:pharmaceutics13122092. [PMID: 34959373 PMCID: PMC8703685 DOI: 10.3390/pharmaceutics13122092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common complication of critical illness and remains a major source of morbidity and mortality in the intensive care unit (ICU). ARDS is characterised by diffuse lung inflammation, epithelial and endothelial deterioration, alveolar–capillary leak and oedema formation, and worsening respiratory failure. The present study aimed to investigate the anti-inflammatory activity of nitric-oxide-releasing dexamethasone derivative NCX-1005 as a potential novel drug for ARDS. Adult rabbits with lavage-induced ARDS were treated with dexamethasone i.v. (0.5 mg/kg; DEX) and nitro-dexamethasone i.v. (0.5 mg/kg, NCX-1005) or were untreated (ARDS). Controls represented healthy ventilated animals. The animals were subsequently oxygen-ventilated for an additional 4 h and respiratory parameters were recorded. Lung oedema, inflammatory cell profile in blood and bronchoalveolar lavage, levels of the cytokines (IL-1β, IL-6, IL-8, TNF-α), and oxidative damage (TBARS, 3NT) in the plasma and lung were evaluated. Nitric oxide-releasing dexamethasone derivative NCX-1005 improved lung function, reduced levels of cytokines, oxidative modifications, and lung oedema formation to similar degrees as dexamethasone. Only NCX-1005 prevented the migration of neutrophils into the lungs compared to dexamethasone. In conclusion, the nitric oxide-releasing dexamethasone derivative NCX-1005 has the potential to be effective drug with anti-inflammatory effect in experimental ARDS.
Collapse
|
10
|
Bick AJ, Louw-du Toit R, Skosana SB, Africander D, Hapgood JP. Pharmacokinetics, metabolism and serum concentrations of progestins used in contraception. Pharmacol Ther 2021; 222:107789. [PMID: 33316287 PMCID: PMC8122039 DOI: 10.1016/j.pharmthera.2020.107789] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
Many different forms of hormonal contraception are used by millions of women worldwide. These contraceptives differ in the dose and type of synthetic progestogenic compound (progestin) used, as well as the route of administration and whether or not they contain estrogenic compounds. There is an increasing awareness that different forms of contraception and different progestins have different side-effect profiles, in particular their cardiovascular effects, effects on reproductive cancers and susceptibility to infectious diseases. There is a need to develop new methods to suit different needs and with minimal risks, especially in under-resourced areas. This requires a better understanding of the pharmacokinetics, metabolism, serum and tissue concentrations of progestins used in contraception as well as the biological activities of progestins and their metabolites via steroid receptors. Here we review the current knowledge on these topics and identify the research gaps. We show that there is a paucity of research on most of these topics for most progestins. We find that major impediments to clear conclusions on these topics include a lack of standardized methodologies, comparisons between non-parallel clinical studies and variability of data on serum concentrations between and within studies. The latter is most likely due, at least in part, to differences in intrinsic characteristics of participants. The review highlights the importance of insight on these topics in order to provide the best contraceptive options to women with minimal risks.
Collapse
Affiliation(s)
- Alexis J Bick
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch 7700, South Africa
| | - Renate Louw-du Toit
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Salndave B Skosana
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch 7700, South Africa
| | - Donita Africander
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Janet P Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch 7700, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
11
|
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
|
12
|
Insights into glucocorticoid responses derived from omics studies. Pharmacol Ther 2020; 218:107674. [PMID: 32910934 DOI: 10.1016/j.pharmthera.2020.107674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 08/20/2020] [Indexed: 12/26/2022]
Abstract
Glucocorticoid drugs are commonly used in the treatment of several conditions, including autoimmune diseases, asthma and cancer. Despite their widespread use and knowledge of biological pathways via which they act, much remains to be learned about the cell type-specific mechanisms of glucocorticoid action and the reasons why patients respond differently to them. In recent years, human and in vitro studies have addressed these questions with genomics, transcriptomics and other omics approaches. Here, we summarize key insights derived from omics studies of glucocorticoid response, and we identify existing knowledge gaps related to mechanisms of glucocorticoid action that future studies can address.
Collapse
|
13
|
Mikolka P, Kosutova P, Kolomaznik M, Topercerova J, Kopincova J, Calkovska A, Mokra D. Effect of different dosages of dexamethasone therapy on lung function and inflammation in an early phase of acute respiratory distress syndrome model. Physiol Res 2020; 68:S253-S263. [PMID: 31928043 DOI: 10.33549/physiolres.934364] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Inflammation associated with acute respiratory distress syndrome (ARDS) can damage the alveolar epithelium and surfactant and worsen the respiratory failure. Glucocorticoids (GC) appear to be a rational therapeutic approach, but the effect is still unclear, especially for early administration and low-dose. In this study we compared two low doses of dexamethasone in early phase of surfactant-depleted model of acute respiratory distress syndrome (ARDS). In the study, lung-lavaged New Zealand rabbits with respiratory failure (PaO(2)<26.7 kPa in FiO(2) 1.0) were treated with intravenous dexamethasone (DEX): 0.5 mg/kg (DEX-0.5) and 1.0 mg/kg (DEX-1.0), or were untreated (ARDS). Animals without ARDS served as controls. Respiratory parameters, lung edema, leukocyte shifts, markers of inflammation and oxidative damage in the plasma and lung were evaluated. Both doses of DEX improved the lung function vs. untreated animals. DEX-1.0 had faster onset with significant improvement in gas exchange and ventilation efficiency vs. DEX-0.5. DEX-1.0 showed a trend to reduce lung neutrophils, local oxidative damage, and levels of TNFalpha, IL-6, IL-8 more effectively than DEX-0.5 vs. ARDS group. Both dosages of dexamethasone significantly improved the lung function and suppressed inflammation in early phase ARDS, while some additional enhancement was observed for higher dose (1 mg/kg) of DEX.
Collapse
Affiliation(s)
- P Mikolka
- Department of Physiology and Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
14
|
Meduri GU, Chrousos GP. General Adaptation in Critical Illness: Glucocorticoid Receptor-alpha Master Regulator of Homeostatic Corrections. Front Endocrinol (Lausanne) 2020; 11:161. [PMID: 32390938 PMCID: PMC7189617 DOI: 10.3389/fendo.2020.00161] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
In critical illness, homeostatic corrections representing the culmination of hundreds of millions of years of evolution, are modulated by the activated glucocorticoid receptor alpha (GRα) and are associated with an enormous bioenergetic and metabolic cost. Appreciation of how homeostatic corrections work and how they evolved provides a conceptual framework to understand the complex pathobiology of critical illness. Emerging literature place the activated GRα at the center of all phases of disease development and resolution, including activation and re-enforcement of innate immunity, downregulation of pro-inflammatory transcription factors, and restoration of anatomy and function. By the time critically ill patients necessitate vital organ support for survival, they have reached near exhaustion or exhaustion of neuroendocrine homeostatic compensation, cell bio-energetic and adaptation functions, and reserves of vital micronutrients. We review how critical illness-related corticosteroid insufficiency, mitochondrial dysfunction/damage, and hypovitaminosis collectively interact to accelerate an anti-homeostatic active process of natural selection. Importantly, the allostatic overload imposed by these homeostatic corrections impacts negatively on both acute and long-term morbidity and mortality. Since the bioenergetic and metabolic reserves to support homeostatic corrections are time-limited, early interventions should be directed at increasing GRα and mitochondria number and function. Present understanding of the activated GC-GRα's role in immunomodulation and disease resolution should be taken into account when re-evaluating how to administer glucocorticoid treatment and co-interventions to improve cellular responsiveness. The activated GRα interdependence with functional mitochondria and three vitamin reserves (B1, C, and D) provides a rationale for co-interventions that include prolonged glucocorticoid treatment in association with rapid correction of hypovitaminosis.
Collapse
Affiliation(s)
- Gianfranco Umberto Meduri
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Memphis Veterans Affairs Medical Center, Memphis, TN, United States
| | - George P. Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
15
|
Matera MG, Rinaldi B, Calzetta L, Rogliani P, Cazzola M. Pharmacokinetics and pharmacodynamics of inhaled corticosteroids for asthma treatment. Pulm Pharmacol Ther 2019; 58:101828. [PMID: 31349002 DOI: 10.1016/j.pupt.2019.101828] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/07/2019] [Accepted: 07/22/2019] [Indexed: 11/27/2022]
Abstract
The differences in the pharmacokinetic (PK) characteristics of inhaled corticosteroids (ICSs) critically influence the profile of each of them, but also the significant differences in glucocorticoid receptor selectivity, potency, and physicochemical properties are critical in defining the pharmacodynamic (PD) profile of an ICS. The PK and PD properties of ICSs used in asthma and the importance of their interrelationship have been reviewed. The differences among the ICSs in PK and PD must be considered when an ICS should be prescribed to an asthmatic patient because a better understanding of the PK/PD interrelationship of ICSs could be important to better fit with the between-patient variability and within-patient repeatability in the response to ICSs that often complicate the therapeutic approach to the asthmatic patient. The role of the device in influencing the PK profile of an ICS must be always considered because it is crucial. Also patient-related factors and disease severity affect pulmonary deposition of ICS.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- University of Campania "Luigi Vanvitelli", Department of Experimental Medicine, Naples, Italy
| | - Barbara Rinaldi
- University of Campania "Luigi Vanvitelli", Department of Experimental Medicine, Naples, Italy
| | - Luigino Calzetta
- University of Rome "Tor Vergata", Department of Experimental Medicine, Rome, Italy
| | - Paola Rogliani
- University of Rome "Tor Vergata", Department of Experimental Medicine, Rome, Italy
| | - Mario Cazzola
- University of Rome "Tor Vergata", Department of Experimental Medicine, Rome, Italy.
| |
Collapse
|
16
|
Zhou W, Chen Z, Li W, Wang Y, Li X, Yu H, Ran P, Liu Z. Systems pharmacology uncovers the mechanisms of anti-asthma herbal medicine intervention (ASHMI) for the prevention of asthma. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
17
|
Gorbacheva AM, Kuprash DV, Mitkin NA. Glucocorticoid Receptor Binding Inhibits an Intronic IL33 Enhancer and is Disrupted by rs4742170 (T) Allele Associated with Specific Wheezing Phenotype in Early Childhood. Int J Mol Sci 2018; 19:ijms19123956. [PMID: 30544846 PMCID: PMC6321062 DOI: 10.3390/ijms19123956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin 33 (IL-33) is a cytokine constitutively expressed by various cells of barrier tissues that contribute to the development of inflammatory immune responses. According to its function as an alarmin secreted by lung and airway epithelium, IL-33 plays a significant role in pathogenesis of allergic disorders. IL-33 is strongly involved in the pathogenesis of asthma, anaphylaxis, allergy and dermatitis, and genetic variations in IL33 locus are associated with increased susceptibility to asthma. Genome-wide association studies have identified risk "T" allele of the single-nucleotide polymorphism rs4742170 located in putative IL33 enhancer area as susceptible variant for development of specific wheezing phenotype in early childhood. Here, we demonstrate that risk "T" rs4742170 allele disrupts binding of glucocorticoid receptor (GR) transcription factor to IL33 putative enhancer. The IL33 promoter/enhancer constructs containing either 4742170 (T) allele or point mutations in the GR-binding site, were significantly more active and did not respond to cortisol in a pulmonary epithelial cell line. At the same time, the constructs containing rs4742170 (C) allele with a functional GR-binding site were less active and further inhibitable by cortisol. The latter effect was GR-dependent as it was completely abolished by GR-specific siRNA. This mechanism may explain the negative effect of the rs4742170 (T) risk allele on the development of wheezing phenotype that strongly correlates with allergic sensitization in childhood.
Collapse
Affiliation(s)
- Alisa M Gorbacheva
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia.
- Biological Faculty, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | - Dmitry V Kuprash
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia.
- Biological Faculty, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | - Nikita A Mitkin
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia.
| |
Collapse
|
18
|
Novel role for receptor dimerization in post-translational processing and turnover of the GRα. Sci Rep 2018; 8:14266. [PMID: 30250038 PMCID: PMC6155283 DOI: 10.1038/s41598-018-32440-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/07/2018] [Indexed: 01/06/2023] Open
Abstract
Glucocorticoids (GCs), acting via the glucocorticoid receptor (GRα), remain the mainstay therapeutic choice for the treatment of inflammation. However, chronic GC use, aside from generating undesirable side-effects, results in GRα down-regulation, often coupled to a decrease in GC-responsiveness, which may culminate in acquired GC resistance. The current study presents evidence for a novel role of the dimerization state of the GRα in mediating GC-mediated GRα turnover. Through comparing the effects of dimerization promoting GCs on down-regulation of a transfected human wild type GRα (hGRwt) or a dimerization deficient GRα mutant (hGRdim), we established that a loss of receptor dimerization restricts GRα turnover, which was supported by the use of the dimerization abrogating Compound A (CpdA), in cells containing endogenous GRα. Moreover, we showed that the dimerization state of the GRα influenced the post-translational processing of the receptor, specifically hyper-phosphorylation at Ser404, which influenced the interaction of GRα with the E3 ligase, FBXW7α, thus hampering receptor turnover via the proteasome. Lastly, the restorative effects of CpdA on the GRα pool, in the presence of Dex, were demonstrated in a combinatorial treatment protocol. These results expand our understanding of factors that contribute to GC-resistance and may be exploited clinically.
Collapse
|
19
|
Rider CF, Carlsten C. Air pollution and resistance to inhaled glucocorticoids: Evidence, mechanisms and gaps to fill. Pharmacol Ther 2018; 194:1-21. [PMID: 30138638 DOI: 10.1016/j.pharmthera.2018.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Substantial evidence indicates that cigarette smoke exposure induces resistance to glucocorticoids, the primary maintenance medication in asthma treatment. Modest evidence also suggests that air pollution may reduce the effectiveness of these critical medications. Cigarette smoke, which has clear parallels with air pollution, has been shown to induce glucocorticoid resistance in asthma and it has been speculated that air pollution may have similar effects. However, the literature on an association of air pollution with glucocorticoid resistance is modest to date. In this review, we detail the evidence for, and against, the effects of air pollution on glucocorticoid effectiveness, focusing on results from epidemiology and controlled human exposure studies. Epidemiological studies indicate a correlation between increased air pollution exposure and worse asthma symptoms. But these studies also show a mix of beneficial and harmful effects of glucocorticoids on spirometry and asthma symptoms, perhaps due to confounding influences, or the induction of glucocorticoid resistance. We describe mechanisms that may contribute to reductions in glucocorticoid responsiveness following air pollution exposure, including changes to phosphorylation or oxidation of the glucocorticoid receptor, repression by cytokines, or inflammatory pathways, and epigenetic effects. Possible interactions between air pollution and respiratory infections are also briefly discussed. Finally, we detail a number of therapies that may boost glucocorticoid effectiveness or reverse resistance in the presence of air pollution, and comment on the beneficial effects of engineering controls, such as air filtration and asthma action plans. We also call attention to the benefits of improved clean air policy on asthma. This review highlights numerous gaps in our knowledge of the interactions between air pollution and glucocorticoids to encourage further research in this area with a view to reducing the harm caused to those with airways disease.
Collapse
Affiliation(s)
- Christopher F Rider
- Respiratory Medicine, Faculty of Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease (COERD), University of British Columbia, Vancouver, BC, Canada.
| | - Chris Carlsten
- Respiratory Medicine, Faculty of Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease (COERD), University of British Columbia, Vancouver, BC, Canada; Institute for Heart and Lung Health, University of British Columbia, Vancouver, BC, Canada; School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
20
|
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 DOI: 10.1124/mol.118.112755] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [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
|
21
|
Taylor RJ, Schlosser RJ, Soler ZM, Mattos JL, Mulligan JK. Glucocorticoid receptor isoform expression in peripheral blood mononuclear leukocytes of patients with chronic rhinosinusitis. Int Forum Allergy Rhinol 2018; 8:10.1002/alr.22120. [PMID: 29719127 PMCID: PMC6214788 DOI: 10.1002/alr.22120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/22/2018] [Accepted: 03/01/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND In several inflammatory disorders, altered peripheral blood mononuclear leukocyte (PBML) glucocorticoid (GC) receptor isoform expression has been associated with GC resistance and disease severity. However, it is unclear if PBML GC receptor isoforms are expressed differentially and are associated with worsened disease severity in chronic rhinosinusitis (CRS). METHODS PBMLs were isolated from control (n = 8), CRS without nasal polyps (CRSsNP) (n = 8), atopic CRS with nasal polyps (CRSwNP) (n = 8), non-atopic CRSwNP (n = 8), and allergic fungal rhinosinusitis (AFRS) (n = 8) patients. Demographics, atopic status, asthmatic status, 22-item Sino-Nasal Outcome Test (SNOT-22) scores, Lund-Kennedy nasal endoscopy scores, Lund-Mackay sinus computed tomography (CT) scores, Kennedy Osteitis scores, and GC utilization 6 months postoperatively were collected. Intracellular immunostaining was then performed for functional GC receptor α (GCRα) and nonfunctional GC receptor β (GCRβ), followed by flow cytometry analysis of geometric mean fluorescent intensity (MFI) and the percentage of cells expressing each GC receptor isoform. RESULTS Compared to controls, each CRS subtype had decreased PBML GCRα and GCRα:GCRβ MFI expression, but no difference in GCRβ expression. Decreasing PBML GCRα in AFRS was associated with increasing Lund-Mackay sinus CT scores (r = -0.880, p =0.004). No significant associations were found between GC receptor isoform expression and other clinical measures. CONCLUSION CRS patients have reduced functional PBML GCRα expression and decreased GCRα:GCRβ compared to controls. Reductions in GCRα in AFRS are associated with worsening Lund-Mackay sinus CT scores. The clinical implications of decreased functional GC receptor expression merits further investigation.
Collapse
Affiliation(s)
- Robert J. Taylor
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
| | - Rodney J. Schlosser
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson VA Medical Center, Charleston, SC
| | - Zachary M. Soler
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
| | - Jose L. Mattos
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
| | - Jennifer K. Mulligan
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
22
|
Papakonstantinou E, Klagas I, Karakiulakis G, Tamm M, Roth M, Stolz D. Glucocorticoids and β 2 -agonists regulate the pathologic metabolism of hyaluronic acid in COPD. Pulm Pharmacol Ther 2018; 48:104-110. [DOI: 10.1016/j.pupt.2017.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 11/28/2022]
|
23
|
Patil RH, Naveen Kumar M, Kiran Kumar KM, Nagesh R, Kavya K, Babu RL, Ramesh GT, Chidananda Sharma S. Dexamethasone inhibits inflammatory response via down regulation of AP-1 transcription factor in human lung epithelial cells. Gene 2017; 645:85-94. [PMID: 29248584 DOI: 10.1016/j.gene.2017.12.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 01/22/2023]
Abstract
The production of inflammatory mediators by epithelial cells in inflammatory lung diseases may represent an important target for the anti-inflammatory effects of glucocorticoids. Activator protein-1 is a major activator of inflammatory genes and has been proposed as a target for inhibition by glucocorticoids. We have used human pulmonary type-II A549 cells to examine the effect of dexamethasone on the phorbol ester (PMA)/Lipopolysaccharide (LPS) induced pro-inflammatory cytokines and AP-1 factors. A549 cells were treated with and without PMA or LPS or dexamethasone and the cell viability and nitric oxide production was measured by MTT assay and Griess reagent respectively. Expression of pro-inflammatory cytokines and AP-1 factors mRNA were measured using semi quantitative RT-PCR. The PMA/LPS treated cells show significant 2-3 fold increase in the mRNA levels of pro-inflammatory cytokines (IL-1β, IL-2, IL-6, IL-8 and TNF-α), cyclo‑oxygenase-2 (COX-2) and specific AP-1 factors (c-Jun, c-Fos and Jun-D). Whereas, pretreatment of cells with dexamethasone significantly inhibited the LPS induced nitric oxide production and PMA/LPS induced mRNAs expression of above pro-inflammatory cytokines, COX-2 and AP-1 factors. Cells treated with dexamethasone alone at both the concentrations inhibit the mRNAs expression of IL-1β, IL-6 and TNF-α compared to control. Our study reveals that dexamethasone decreased the mRNAs expression of c-Jun and c-Fos available for AP-1 formation suggested that AP-1 is the probable key transcription factor involved in the anti-inflammatory activity of dexamethasone. This may be an important molecular mechanism of steroid action in asthma and other chronic inflammatory lung diseases which may be useful for treatment of lung inflammatory diseases.
Collapse
Affiliation(s)
- Rajeshwari H Patil
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India; Department of Biotechnology, The Oxford College of Science, HSR Layout, Bengaluru 560102, Karnataka, India.
| | - M Naveen Kumar
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India
| | - K M Kiran Kumar
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India
| | - Rashmi Nagesh
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India
| | - K Kavya
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India
| | - R L Babu
- Department of Bioinformatics and Biotechnology, Karnataka State Women's University, Jnana Shakthi Campus, Vijayapura 586 108, Karnataka, India; Department of Biology and Center for Biotechnology and Biomedical Sciences, Norfolk State University, Norfolk, VA, USA
| | - Govindarajan T Ramesh
- Department of Biology and Center for Biotechnology and Biomedical Sciences, Norfolk State University, Norfolk, VA, USA
| | - S Chidananda Sharma
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru 560 056, Karnataka, India
| |
Collapse
|
24
|
Moosavi SM, Prabhala P, Ammit AJ. Role and regulation of MKP-1 in airway inflammation. Respir Res 2017; 18:154. [PMID: 28797290 PMCID: PMC5554001 DOI: 10.1186/s12931-017-0637-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/04/2017] [Indexed: 12/18/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) is a protein with anti-inflammatory properties and the archetypal member of the dual-specificity phosphatases (DUSPs) family that have emerged over the past decade as playing an instrumental role in the regulation of airway inflammation. Not only does MKP-1 serve a critical role as a negative feedback effector, controlling the extent and duration of pro-inflammatory MAPK signalling in airway cells, upregulation of this endogenous phosphatase has also emerged as being one of the key cellular mechanism responsible for the beneficial actions of clinically-used respiratory medicines, including β2-agonists, phosphodiesterase inhibitors and corticosteroids. Herein, we review the role and regulation of MKP-1 in the context of airway inflammation. We initially outline the structure and biochemistry of MKP-1 and summarise the multi-layered molecular mechanisms responsible for MKP-1 production more generally. We then focus in on some of the key in vitro studies in cell types relevant to airway disease that explain how MKP-1 can be regulated in airway inflammation at the transcriptional, post-translation and post-translational level. And finally, we address some of the potential challenges with MKP-1 upregulation that need to be explored further to fully exploit the potential of MKP-1 to repress airway inflammation in chronic respiratory disease.
Collapse
Affiliation(s)
- Seyed M Moosavi
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Pavan Prabhala
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Alaina J Ammit
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia. .,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
25
|
Yoo H, Kang M, Pyo S, Chae HS, Ryu KH, Kim J, Chin YW. SKI3301, a purified herbal extract from Sophora tonkinensis, inhibited airway inflammation and bronchospasm in allergic asthma animal models in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:298-305. [PMID: 28506902 DOI: 10.1016/j.jep.2017.05.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/30/2017] [Accepted: 05/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sophora tonkinensis (Leguminosae, ST) is a traditional herbal plant in Korea and China. Its roots and rhizomes have been used to dissipate heat, to clear toxic material and to treat acute pharyngolaryngeal infections and sore throats. AIM OF STUDY In this study, we tried to investigate the anti-inflammatory and anti-asthmatic effects of a purified extract (SKI3301) from Sophora tonkinensis using in vitro enzyme assay models and ovalbumin (OVA)-induced asthma animal models. MATERIALS AND METHODS The effect of SKI3301 on pro-inflammatory enzymes such as 5-lipoxygenase, phosphodiesterase 3 & 4, and thromboxane synthase was assayed in vitro. BALB/c mice were sensitized with OVA/Alum ip injection and nebulized with OVA to induce airway inflammation. Bronchoalveolar lavage (BAL) fluid was collected and analyzed for leukocytes infiltration and IL-5 production along with lung histopathology. Guinea pigs passively sensitized with anti-OVA antiserum were used to investigate the effect of SKI3301 on bronchospasm in vitro and in vivo. RESULTS SKI3301 potently inhibited the activities of 5-lipoxygenase, phosphodiesterase 3 & 4, and thromboxane synthase. Orally administered SKI3301 attenuated the total leukocytes and eosinophil infiltration and IL-5 level in BAL fluids. Histopathological changes associated with lung inflammation were also reduced by SKI3301. SKI3301 inhibited OVA-induced contraction of isolated trachea from sensitized guinea pigs. SKI3301 also protected OVA-induced bronchoconstriction in the sensitized guinea pigs. Maackiain, one of 3 major components of SKI3301, was effective in inhibiting 5-lipoxygenase and OVA-induced airway inflammation. CONCLUSION In this study, SKI3301 potently inhibited pro-inflammatory enzymes and attenuated OVA-induced bronchospasm in animal model of allergic asthma. These results suggest that SKI3301 may have therapeutic potential for allergic asthma.
Collapse
Affiliation(s)
- Hunseung Yoo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08862, Republic of Korea; New Drug Preclinical & Analytical Team, Life Science R&D Center, SK Chemicals, 310 Pangyo-ro, 463-400 Republic of Korea
| | - Minseok Kang
- New Drug Preclinical & Analytical Team, Life Science R&D Center, SK Chemicals, 310 Pangyo-ro, 463-400 Republic of Korea
| | - Sungsoo Pyo
- New Drug Preclinical & Analytical Team, Life Science R&D Center, SK Chemicals, 310 Pangyo-ro, 463-400 Republic of Korea
| | - Hee-Sung Chae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Keun Ho Ryu
- New Drug Preclinical & Analytical Team, Life Science R&D Center, SK Chemicals, 310 Pangyo-ro, 463-400 Republic of Korea
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08862, Republic of Korea.
| | - Young-Won Chin
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| |
Collapse
|
26
|
Mikolka P, Kopincová J, Košútová P, Čierny D, Čalkovská A, Mokrá D. Lung inflammatory and oxidative alterations after exogenous surfactant therapy fortified with budesonide in rabbit model of meconium aspiration syndrome. Physiol Res 2017; 65:S653-S662. [PMID: 28006947 DOI: 10.33549/physiolres.933529] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Meconium aspiration syndrome (MAS) triggers inflammatory and oxidative pathways which can inactivate both pulmonary surfactant and therapeutically given exogenous surfactant. Glucocorticoid budesonide added to exogenous surfactant can inhibit inflammation and thereby enhance treatment efficacy. Neonatal meconium (25 mg/ml, 4 ml/kg) was administered intratracheally (i.t.) to rabbits. When the MAS model was prepared, animals were treated with budesonide i.t. (Pulmicort, 0.25 mg/kg, M+B); with surfactant lung lavage (Curosurf®, 10 ml/kg, 5 mg phospholipids/ml, M+S) followed by undiluted Curosurf® i.t. (100 mg phospholipids/kg); with combination of budesonide and surfactant (M+S+B); or were untreated (M); or served as controls with saline i.t. instead of meconium (C). Animals were oxygen-ventilated for additional 5 h. Cell counts in the blood and bronchoalveolar lavage fluid (BAL), lung edema formation (wet/dry weight ratio), oxidative damage of lipids/ proteins and inflammatory expression profiles (IL-2, IL-6, IL-13, TNF-alpha) in the lung homogenate and plasma were determined. Combined surfactant+budesonide therapy was the most effective in reduction of neutrophil counts in BAL, oxidative damage, levels and mRNA expression of cytokines in the lung, and lung edema formation compared to untreated animals. Curosurf fortified with budesonide mitigated lung inflammation and oxidative modifications what indicate the perspectives of this treatment combination for MAS therapy.
Collapse
Affiliation(s)
- P Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
27
|
Rigo LA, Carvalho-Wodarz CS, Pohlmann AR, Guterres SS, Schneider-Daum N, Lehr CM, Beck RCR. Nanoencapsulation of a glucocorticoid improves barrier function and anti-inflammatory effect on monolayers of pulmonary epithelial cell lines. Eur J Pharm Biopharm 2017; 119:1-10. [PMID: 28512018 DOI: 10.1016/j.ejpb.2017.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 05/08/2017] [Accepted: 05/12/2017] [Indexed: 01/15/2023]
Abstract
The anti-inflammatory effect of polymeric deflazacort nanocapsules (NC-DFZ) was investigated, and possible improvement of epithelial barrier function using filter grown monolayers of Calu-3 cells was assessed. NC prepared from poly(ε-caprolactone) (PCL) had a mean size around 200nm, slightly negative zeta potential (∼-8mV), and low polydispersity index (<0.10). Encapsulation of DFZ had an efficiency of 85%. No cytotoxic effects were observed at particle concentration of 9.85×1011NC/ml, which was therefore chosen to evaluate the effect of NC-DFZ at 1% (w/v) of PCL and 0.5% (w/v) of DFZ on the epithelial barrier function of Calu-3 monolayers. Nanoencapsulated drug at 0.5% (w/v) increased transepithelial electrical resistance and decreased permeability of the paracellular marker sodium fluorescein, while non-encapsulated DFZ failed to improve these parameters. Moreover, NC-DFZ reduced the lipopolysaccharide (LPS) mediated secretion of the inflammatory marker IL-8. In vitro dissolution testing revealed controlled release of DFZ from nanocapsules, which may explain the improved effect of DFZ on the cells. These data suggest that nanoencapsulation of pulmonary delivered corticosteroids could be advantageous for the treatment of inflammatory conditions, such as asthma and chronic obstructive pulmonary diseases.
Collapse
Affiliation(s)
- Lucas A Rigo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Cristiane S Carvalho-Wodarz
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), University Campus, Building E8.1, D-66123 Saarbrücken, Germany
| | - Adriana R Pohlmann
- Departamento de Química Orgânica, Instituto de Química, UFRGS, Av. Bento Gonçalves 9500, 91501-970, Brazil
| | - Silvia S Guterres
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Nicole Schneider-Daum
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), University Campus, Building E8.1, D-66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), University Campus, Building E8.1, D-66123 Saarbrücken, Germany
| | - Ruy C R Beck
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga, 2752, 90610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
28
|
Kim HS, Won S, Lee EK, Chun YH, Yoon JS, Kim JT, Kim HH. Effect of Proparacaine in a Mouse Model of Allergic Rhinitis. Clin Exp Otorhinolaryngol 2017; 10:325-331. [PMID: 28449552 PMCID: PMC5678040 DOI: 10.21053/ceo.2017.00101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/27/2017] [Accepted: 03/31/2017] [Indexed: 01/05/2023] Open
Abstract
Objectives Lidocaine, a local anaesthetic is a treatment option in uncontrolled asthma due to its immunomodulatory effects. In the present study, proparacaine (PPC), a derivative of lidocaine was examined for its therapeutic application in a mouse model of allergic rhinitis. Methods The mice were grouped into 4 groups: control group, allergic rhinitis (AR) group, ciclesonide (CIC) group, and PPC group. Nasal symptom scores, eosinophil counts, goblet cell counts, and mast cells counts in the nasal mucosa were measured. Serum ovalbumin (OVA)-specific immunoglobulin (Ig) E, OVA-specific IgG1, OVA-specific IgG2a, interleukin (IL)-4, IL-5, and cortisol levels were measured. Results Intranasal administration of PPC significantly decreased nasal symptoms, number of eosinophils, goblet cells, and mast cells in the lamina propria of the nasal mucosa. Serum OVA-specific IgE, OVA-specific IgG1, OVA-specific IgG2a was significantly higher in the AR compared with the control group. Serum level of IL-4 was significantly lower in the CIC group and PPC group in comparison with AR group. Serum IL-5 showed no significant difference among all groups. No significant difference in serum cortisol levels was observed among the 4 groups. Conclusion PPC appears to have a therapeutic potential in treatment of allergic rhinitis in a mouse model by reducing eosinophil, goblet cell, and mast cell infiltration in the nasal mucosa.
Collapse
Affiliation(s)
- Hwan Soo Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sulmui Won
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eu Kyoung Lee
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon Hong Chun
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong-Seo Yoon
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Tack Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun Hee Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
29
|
Keränen T, Moilanen E, Korhonen R. Suppression of cytokine production by glucocorticoids is mediated by MKP-1 in human lung epithelial cells. Inflamm Res 2017; 66:441-449. [PMID: 28299397 DOI: 10.1007/s00011-017-1028-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 01/12/2017] [Accepted: 02/15/2017] [Indexed: 11/30/2022] Open
Abstract
Mitogen-activated protein kinase phosphatase 1 (MKP-1) expression is induced by inflammatory factors and serves as an endogenous p38 MAPK suppressor to limit inflammatory response. Glucocorticoids are very effective anti-inflammatory drugs and they are used for the treatment of many inflammatory diseases, such as asthma and COPD. We investigated the role of MKP-1 in the inhibition of cytokine production by dexamethasone in human A549 bronchial epithelial cells. We found that dexamethasone increased MKP-1 expression, inhibited p38 MAPK phosphorylation, and suppressed TNF and MIP-3α production in A549 cells. Interestingly, the suppression of p38 MAPK phosphorylation and the inhibition of TNF expression by dexamethasone were attenuated in cells, where MKP-1 expression was silenced by siRNA. In conclusion, these data suggest that dexamethasone increases MKP-1 expression and this results in the suppression of p38 MAPK signaling leading to the inhibition of cytokine production in human bronchial epithelial cells. These results point to the role of MKP-1 as an important factor in the therapeutic effects of glucocorticoids in the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Tiina Keränen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland
| | - Riku Korhonen
- The Immunopharmacology Research Group, University of Tampere School of Medicine, and Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
Shah S, King EM, Mostafa MM, Altonsy MO, Newton R. DUSP1 Maintains IRF1 and Leads to Increased Expression of IRF1-dependent Genes: A MECHANISM PROMOTING GLUCOCORTICOID INSENSITIVITY. J Biol Chem 2016; 291:21802-21816. [PMID: 27551049 DOI: 10.1074/jbc.m116.728964] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/19/2016] [Indexed: 11/06/2022] Open
Abstract
Although the mitogen-activated protein kinase (MAPK) phosphatase, DUSP1, mediates dexamethasone-induced repression of MAPKs, 14 of 46 interleukin-1β (IL1B)-induced mRNAs were significantly enhanced by DUSP1 overexpression in pulmonary A549 cells. These include the interferon regulatory factor, IRF1, and the chemokine, CXCL10. Of these, DUSP1-enhanced mRNAs, 10 including CXCL10, were IRF1-dependent. MAPK inhibitors and DUSP1 overexpression prolonged IRF1 expression by elevating transcription and increasing IRF1 mRNA and protein stability. Conversely, DUSP1 silencing increased IL1B-induced MAPK phosphorylation while significantly reducing IRF1 protein expression at 4 h. This confirms a regulatory network whereby DUSP1 switches off MAPKs to maintain IRF1 expression. There was no repression of IRF1 expression by dexamethasone in primary human bronchial epithelial cells, and in A549 cells IL1B-induced IRF1 protein was only modestly and transiently repressed. Although dexamethasone did not repress IL1B-induced IRF1 protein expression at 4-6 h, silencing of IL1B plus dexamethasone-induced DUSP1 significantly reduced IRF1 expression. IL1B-induced expression of CXCL10 was largely insensitive to dexamethasone, whereas other DUSP1-enhanced, IRF1-dependent mRNAs showed various degrees of repression. With IL1B plus dexamethasone, CXCL10 expression was also IRF1-dependent, and expression was reduced by DUSP1 silencing. Thus, IL1B plus dexamethasone-induced DUSP1 maintains expression of IRF1 and the IRF1-dependent gene, CXCL10. This is supported by chromatin immunoprecipitation showing IRF1 recruitment to be essentially unaffected by dexamethasone at the CXCL10 promoter or at the promoters of more highly repressed IRF1-dependent genes. Since IRF1-dependent genes, such as CXCL10, are central to host defense, these data may help explain the reduced effectiveness of glucocorticoids during asthma exacerbations.
Collapse
Affiliation(s)
- Suharsh Shah
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Elizabeth M King
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Mahmoud M Mostafa
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| | - Mohammed O Altonsy
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and.,Department of Zoology, Sohag University, Sohag 825224, Egypt
| | - Robert Newton
- From the Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada T2N 4Z6 and
| |
Collapse
|
34
|
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
|
35
|
Hapgood JP, Avenant C, Moliki JM. Glucocorticoid-independent modulation of GR activity: Implications for immunotherapy. Pharmacol Ther 2016; 165:93-113. [PMID: 27288728 DOI: 10.1016/j.pharmthera.2016.06.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Pharmacological doses of glucocorticoids (GCs), acting via the glucocorticoid receptor (GR) to repress inflammation and immune function, remain the most effective therapy in the treatment of inflammatory and immune diseases. Since many patients on GC therapy exhibit GC resistance and severe side-effects, much research is focused on developing more selective GCs and combination therapies, with greater anti-inflammatory potency. GCs mediate their classical genomic transcriptional effects by binding to the cytoplasmic GR, followed by nuclear translocation and modulation of transcription of target genes by direct DNA binding of the GR or its tethering to other transcription factors. Recent evidence suggests, however, that the responses mediated by the GR are much more complex and involve multiple parallel mechanisms integrating simultaneous signals from other receptors, both in the absence and presence of GCs, to shift the sensitivity of a target cell to GCs. The level of cellular stress, immune activation status, or the cell cycle phase may be crucial for determining GC sensitivity and GC responsiveness as well as subcellular localization of the GR and GR levels. Central to the development of new drugs that target GR signaling alone or as add-on therapies, is an in-depth understanding of the molecular mechanisms of GC-independent GR desensitization, priming and activation of the unliganded GR, as well as synergy and cross-talk with other signaling pathways. This review will discuss the information currently available on these topics and their relevance to immunotherapy, as well as identify unanswered questions and future areas of research.
Collapse
Affiliation(s)
- Janet P Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch, 7700, South Africa.
| | - Chanel Avenant
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch, 7700, South Africa
| | - Johnson M Moliki
- Department of Molecular and Cell Biology, University of Cape Town, Private Bag X3, Rondebosch, 7700, South Africa
| |
Collapse
|
36
|
8,9-Dehydrohispanolone-15,16-lactol diterpene prevents LPS-triggered inflammatory responses by inhibiting endothelial activation. Biochem J 2016; 473:2061-71. [PMID: 27154204 DOI: 10.1042/bcj20160343] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/06/2016] [Indexed: 02/07/2023]
Abstract
Endothelial activation contributes to lung inflammatory disorders by inducing leucocyte recruitment to pulmonary parenchyma. Consequently, vascular-targeted therapies constitute promising strategies for the treatment of inflammatory pathologies. In the present study, we evaluated the effect of 8,9-dehydrohispanolone-15,16-lactol diterpene (DT) on lung endothelium during inflammation. Lung endothelial cells pre-treated with DT and activated with lipopolysaccharide (LPS) or tumour necrosis factor-α (TNF-α) exhibited reduced expression of the pro-inflammatory cytokines Cxcl10, Ccl5 and Cxcl1, whereas the anti-inflammatory molecules IL1r2 and IL-10 were induced. Consistent with this result, DT pre-treatment inhibited nuclear factor κB (NF-κB) nuclear translocation, by interfering with IκBα phosphorylation, and consequently NF-κB transcriptional activity in endothelium activated by LPS or TNF-α. Furthermore, DT, probably through p38 signalling, induced transcriptional activation of genes containing activator protein 1 (AP-1)-binding elements. Inhibition of p38 prevented IL1r2 mRNA expression in endothelium incubated with DT alone or in combination with LPS or TNF-α. Accordingly, conditioned medium (CM) from these cells failed to stimulate leucocytes as measured by a reduction in adhesive ability of the leucocyte cell line J774 to fibronectin (FN). Additionally, DT reduced the expression of the endothelial adhesion molecules E-selectin, vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) after activation. Similarly, expression of VCAM-1 and ICAM-1 molecules on the lung endothelial layer of C57/BL6 mice pre-treated with DT and challenged with LPS were unchanged. Finally, inhibition of vascular adhesion molecule expression by DT decreased the interaction of J774 cells with lung endothelial cells in an inflammatory environment. Our findings establish DT as a novel endothelial inhibitor for the treatment of inflammatory-related diseases triggered by Gram-negative bacteria or by the associated cytokine TNF-α.
Collapse
|
37
|
Ingawale DK, Mandlik SK, Patel SS. An emphasis on molecular mechanisms of anti-inflammatory effects and glucocorticoid resistance. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2015; 12:1-13. [PMID: 25503867 DOI: 10.1515/jcim-2014-0051] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 11/16/2014] [Indexed: 11/15/2022]
Abstract
Glucocorticoids (GC) are universally accepted agents for the treatment of anti-inflammatory and immunosuppressive disorders. They are used in the treatment of rheumatic diseases and various inflammatory diseases such as allergy, asthma and sepsis. They bind with GC receptor (GR) and form GC-GR complex with the receptor and exert their actions. On activation the GC-GR complex up-regulates the expression of nucleus anti-inflammatory proteins called as transactivation and down-regulates the expression of cytoplasmic pro-inflammatory proteins called as transrepression. It has been observed that transactivation mechanisms are notorious for side effects and transrepressive mechanisms are identified for beneficial anti-inflammatory effects of GC therapy. GC hampers the function of numerous inflammatory mediators such as cytokines, chemokines, adhesion molecules, arachidonic acid metabolites, release of platelet-activating factor (PAF), inflammatory peptides and enzyme modulation involved in the process of inflammation. The GC resistance is a serious therapeutic problem and limits the therapeutic response of GC in chronic inflammatory patients. It has been observed that the GC resistance can be attributed to cellular microenvironment changes, as a consequence of chronic inflammation. Various other factors responsible for resistance have been identified, including alterations in both GR-dependent and GR-independent signaling pathways of cytokine action, hypoxia, oxidative stress, allergen exposure and serum-derived factors. The present review enumerates various aspects of inflammation such as use of GC for treatment of inflammation and its mechanism of action. Molecular mechanisms of anti-inflammatory action of GC and GC resistance, alternative anti-inflammatory treatments and new strategy for reversing the GC resistance have also been discussed.
Collapse
|
38
|
Pelaia G, Muzzio CC, Vatrella A, Maselli R, Magnoni MS, Rizzi A. Pharmacological basis and scientific rationale underlying the targeted use of inhaled corticosteroid/long-acting β2-adrenergic agonist combinations in chronic obstructive pulmonary disease treatment. Expert Opin Pharmacother 2015; 16:2009-21. [DOI: 10.1517/14656566.2015.1070826] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
39
|
Liu X, Feng J, Sun X, Li Y, Duan G. Three-layer structure graphene/mesoporous silica composites incorporated with C8-modified interior pore-walls for residue analysis of glucocorticoids in milk by liquid chromatography–tandem mass spectrometry. Anal Chim Acta 2015; 884:61-9. [DOI: 10.1016/j.aca.2015.05.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 01/29/2023]
|
40
|
Joshi T, Johnson M, Newton R, Giembycz MA. The long-acting β2 -adrenoceptor agonist, indacaterol, enhances glucocorticoid receptor-mediated transcription in human airway epithelial cells in a gene- and agonist-dependent manner. Br J Pharmacol 2015; 172:2634-53. [PMID: 25598440 DOI: 10.1111/bph.13087] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/11/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhaled glucocorticoid (ICS)/long-acting β2 -adrenoceptor agonist (LABA) combination therapy is a recommended treatment option for patients with moderate/severe asthma in whom adequate control cannot be achieved by an ICS alone. Previously, we discovered that LABAs can augment dexamethasone-inducible gene expression and proposed that this effect may explain how these two drugs interact to deliver superior clinical benefit. Herein, we extended that observation by analysing, pharmacodynamically, the effect of the LABA, indacaterol, on glucocorticoid receptor (GR)-mediated gene transcription induced by seven ligands with intrinsic activity values that span the spectrum of full agonism to antagonism. EXPERIMENTAL APPROACH BEAS-2B human airway epithelial cells stably transfected with a 2× glucocorticoid response element luciferase reporter were used to model gene transcription together with an analysis of several glucocorticoid-inducible genes. KEY RESULTS Indacaterol augmented glucocorticoid-induced reporter activation in a manner that was positively related to the intrinsic activity of the GR agonist. This effect was demonstrated by an increase in response maxima without a change in GR agonist affinity or efficacy. Indacaterol also enhanced glucocorticoid-inducible gene expression. However, the magnitude of this effect was dependent on both the GR agonist and the gene of interest. CONCLUSIONS AND IMPLICATIONS These data suggest that indacaterol activates a molecular rheostat, which increases the transcriptional competency of GR in an agonist- and gene-dependent manner without apparently changing the relationship between fractional GR occupancy and response. These findings provide a platform to rationally design ICS/LABA combination therapy that is based on the generation of agonist-dependent gene expression profiles in target and off-target tissues.
Collapse
Affiliation(s)
- T Joshi
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
41
|
Gauvreau GM, Boulet LP, Leigh R, Cockcroft DW, Killian KJ, Davis BE, Deschesnes F, Watson RM, Swystun V, Mårdh CK, Wessman P, Jorup C, Aurivillius M, O'Byrne PM. A nonsteroidal glucocorticoid receptor agonist inhibits allergen-induced late asthmatic responses. Am J Respir Crit Care Med 2015; 191:161-7. [PMID: 25473939 DOI: 10.1164/rccm.201404-0623oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Effective antiinflammatory therapies are needed for the treatment of asthma, but preferably without the systemic adverse effects of glucocorticosteroids. OBJECTIVES We evaluated the effect of an inhaled nonsteroidal glucocorticoid receptor agonist, AZD5423, on allergen-induced responses. METHODS Twenty subjects with mild allergic asthma were randomized to receive 7 days of treatment with nebulized AZD5423 (75 or 300 μg) once daily, budesonide 200 μg twice daily via Turbuhaler, or placebo in a double-blind, four-period, crossover design study. Allergen challenge was performed on Day 6. MEASUREMENTS AND MAIN RESULTS FEV1 was measured repeatedly for 7 hours after allergen challenge for early and late asthmatic responses. Sputum inflammatory cells was measured before and at 7 and 24 hours after allergen challenge, and methacholine airway responsiveness was measured before and 24 hours after allergen challenge. AZD5423 significantly attenuated the fall in FEV1 during the late asthmatic response (both doses led to an 8.7% fall) versus placebo (14% fall) (P < 0.05) with no effect of budesonide (12.5% fall) versus placebo (P > 0.05). There was no effect on the fall in FEV1 during early asthmatic response. AZD5423 300 and 75 μg significantly attenuated allergen-induced sputum eosinophilia by 63 and 61% at 7 hours, respectively, and by 46 and 34% at 24 hours after allergen challenge, respectively, versus placebo (all P < 0.05). Budesonide did not reduce allergen-induced sputum eosinophilia versus placebo. AZD5423 at 300 μg significantly attenuated allergen-induced airway hyperresponsiveness at 24 hours after allergen challenge versus placebo (P < 0.05). Both doses of AZD5423 were well tolerated. CONCLUSIONS Seven-day treatment with inhalation of the nonsteroidal glucocorticoid receptor agonist AZD5423 effectively reduced allergen-induced responses in subjects with mild allergic asthma. Clinical trial registered with www.clinicaltrials.gov (NCT01225549).
Collapse
Affiliation(s)
- Gail M Gauvreau
- 1 Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Joshi T, Johnson M, Newton R, Giembycz M. An analysis of glucocorticoid receptor-mediated gene expression in BEAS-2B human airway epithelial cells identifies distinct, ligand-directed, transcription profiles with implications for asthma therapeutics. Br J Pharmacol 2015; 172:1360-78. [PMID: 25393397 PMCID: PMC4337707 DOI: 10.1111/bph.13014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/08/2014] [Accepted: 11/05/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE International asthma guidelines recommend that inhaled glucocorticoids be used as a monotherapy in all patients with mild to moderate disease because of their ability to suppress airways inflammation. Current evidence suggests that the therapeutic benefit of glucocorticoids is due to the transactivation and transrepression of anti-inflammatory and pro-inflammatory genes respectively. However, the extent to which clinically relevant glucocorticoids are equivalent in their ability to modulate gene expression is unclear. EXPERIMENTAL APPROACH A pharmacodynamics investigation of glucocorticoid receptor (GR)-mediated gene transactivation in BEAS-2B human airway epithelial cells was performed using a glucocorticoid response element luciferase reporter coupled with an analysis of glucocorticoid-inducible genes encoding proteins with anti-inflammatory and adverse-effect potential. KEY RESULTS Using transactivation as a functionally relevant output, a given glucocorticoid displayed a unique, gene expression 'fingerprint' where intrinsic efficacy and GR density were essential determinants. We showed that depending on the gene selected for analysis, a given glucocorticoid can behave as an antagonist, partial agonist, full agonist or even 'super agonist'. In the likely event that different, tissue-dependent gene expression profiles are reproduced in vivo, then the anti-inflammatory and adverse-effect potential of many glucocorticoids currently available as asthma therapeutics may not be equivalent. CONCLUSIONS AND IMPLICATIONS The generation of gene expression 'fingerprints' in target and off-target human tissues could assist the rational design of GR agonists with improved therapeutic ratios. This approach could identify compounds that are useful in the management of severe asthma and other inflammatory disorders where systemic exposure is desirable.
Collapse
Affiliation(s)
- T Joshi
- Airways Inflammation Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - M Johnson
- GlaxoSmithKline Research and DevelopmentUxbridge, Middlesex, UK
| | - R Newton
- Department of Cell Biology and Anatomy, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - M Giembycz
- Airways Inflammation Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| |
Collapse
|
43
|
Giembycz MA, Newton R. Potential mechanisms to explain how LABAs and PDE4 inhibitors enhance the clinical efficacy of glucocorticoids in inflammatory lung diseases. F1000PRIME REPORTS 2015; 7:16. [PMID: 25750734 PMCID: PMC4335793 DOI: 10.12703/p7-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inhaled glucocorticoids acting via the glucocorticoid receptor are a mainstay treatment option for individuals with asthma. There is a consensus that the remedial actions of inhaled glucocorticoids are due to their ability to suppress inflammation by modulating gene expression. While inhaled glucocorticoids are generally effective in asthma, there are subjects with moderate-to-severe disease in whom inhaled glucocorticoids fail to provide adequate control. For these individuals, asthma guidelines recommend that a long-acting β2-adrenoceptor agonist (LABA) be administered concurrently with an inhaled glucocorticoid. This so-called “combination therapy” is often effective and clinically superior to the inhaled glucocorticoid alone, irrespective of dose. LABAs, and another class of drug known as phosphodiesterase 4 (PDE4) inhibitors, may also enhance the efficacy of inhaled glucocorticoids in chronic obstructive pulmonary disease (COPD). In both conditions, these drugs are believed to work by elevating the concentration of cyclic adenosine-3',5'-monophosphate (cAMP) in target cells and tissues. Despite the success of inhaled glucocorticoid/LABA combination therapy, it remains unclear how an increase in cAMP enhances the clinical efficacy of an inhaled glucocorticoid. In this report, we provide a state-of-the-art appraisal, including unresolved and controversial issues, of how cAMP-elevating drugs and inhaled glucocorticoids interact at a molecular level to deliver enhanced anti-inflammatory benefit over inhaled glucocorticoid monotherapy. We also speculate on ways to further exploit this desirable interaction. Critical discussion of how these two drug classes regulate gene transcription, often in a synergistic manner, is a particular focus. Indeed, because interplay between glucocorticoid receptor and cAMP signaling pathways may contribute to the superiority of inhaled glucocorticoid/LABA combination therapy, understanding this interaction may provide a logical framework to rationally design these multicomponent therapeutics that was not previously possible.
Collapse
Affiliation(s)
- Mark A. Giembycz
- Department of Physiology & Pharmacology, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary3820 Hospital Drive NW, Calgary, AlbertaCanada T2N 1N4
| | - Robert Newton
- Department of Cell Biology & Anatomy, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary3820 Hospital Drive NW, Calgary, AlbertaCanada T2N 1N4
| |
Collapse
|
44
|
Rider CF, Shah S, Miller-Larsson A, Giembycz MA, Newton R. Cytokine-induced loss of glucocorticoid function: effect of kinase inhibitors, long-acting β(2)-adrenoceptor [corrected] agonist and glucocorticoid receptor ligands. PLoS One 2015; 10:e0116773. [PMID: 25625944 PMCID: PMC4308083 DOI: 10.1371/journal.pone.0116773] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/13/2014] [Indexed: 11/19/2022] Open
Abstract
Acting on the glucocorticoid receptor (NR3C1), glucocorticoids are widely used to treat inflammatory diseases. However, glucocorticoid resistance often leads to suboptimal asthma control. Since glucocorticoid-induced gene expression contributes to glucocorticoid activity, the aim of this study was to use a 2 × glucocorticoid response element (GRE) reporter and glucocorticoid-induced gene expression to investigate approaches to combat cytokine-induced glucocorticoid resistance. Pre-treatment with tumor necrosis factor-α (TNF) or interleukin-1β inhibited dexamethasone-induced mRNA expression of the putative anti-inflammatory genes RGS2 and TSC22D3, or just TSC22D3, in primary human airway epithelial and smooth muscle cells, respectively. Dexamethasone-induced DUSP1 mRNA was unaffected. In human bronchial epithelial BEAS-2B cells, dexamethasone-induced TSC22D3 and CDKN1C expression (at 6 h) was reduced by TNF pre-treatment, whereas DUSP1 and RGS2 mRNAs were unaffected. TNF pre-treatment also reduced dexamethasone-dependent 2×GRE reporter activation. This was partially reversed by PS-1145 and c-jun N-terminal kinase (JNK) inhibitor VIII, inhibitors of IKK2 and JNK, respectively. However, neither inhibitor affected TNF-dependent loss of dexamethasone-induced CDKN1C or TSC22D3 mRNA. Similarly, inhibitors of the extracellular signal-regulated kinase, p38, phosphoinositide 3-kinase or protein kinase C pathways failed to attenuate TNF-dependent repression of the 2×GRE reporter. Fluticasone furoate, fluticasone propionate and budesonide were full agonists relative to dexamethasone, while GSK9027, RU24858, des-ciclesonide and GW870086X were partial agonists on the 2×GRE reporter. TNF reduced reporter activity in proportion with agonist efficacy. Full and partial agonists showed various degrees of agonism on RGS2 and TSC22D3 expression, but were equally effective at inducing CDKN1C and DUSP1, and did not affect the repression of CDKN1C or TSC22D3 expression by TNF. Finally, formoterol-enhanced 2×GRE reporter activity was also proportional to agonist efficacy and functionally reversed repression by TNF. As similar effects were apparent on glucocorticoid-induced gene expression, the most effective strategy to overcome glucocorticoid resistance in this model was addition of formoterol to high efficacy NR3C1 agonists.
Collapse
Affiliation(s)
- Christopher F. Rider
- Airways Inflammation Research Group, Snyder Institute of Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Suharsh Shah
- Airways Inflammation Research Group, Snyder Institute of Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Mark A. Giembycz
- Airways Inflammation Research Group, Snyder Institute of Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Airways Inflammation Research Group, Snyder Institute of Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
45
|
Donnenfeld ED. Current trends in postsurgical management of ocular inflammation following cataract surgery. EXPERT REVIEW OF OPHTHALMOLOGY 2015. [DOI: 10.1586/17469899.2015.996548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
BinMahfouz H, Borthakur B, Yan D, George T, Giembycz MA, Newton R. Superiority of combined phosphodiesterase PDE3/PDE4 inhibition over PDE4 inhibition alone on glucocorticoid- and long-acting β2-adrenoceptor agonist-induced gene expression in human airway epithelial cells. Mol Pharmacol 2014; 87:64-76. [PMID: 25324049 DOI: 10.1124/mol.114.093393] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoids, also known as corticosteroids, induce effector gene transcription as a part of their anti-inflammatory mechanisms of action. Such genomic effects can be significantly enhanced by long-acting β2-adrenoceptor agonists (LABAs) and may contribute to the clinical superiority of inhaled corticosteroid (ICS)/LABA combinations in asthma and chronic obstructive pulmonary disease (COPD) over ICSs alone. Using models of cAMP- and glucocorticoid-induced transcription in human bronchial epithelial BEAS-2B cells, we show that combining inhibitors of phosphodiesterase (PDE) 3 and PDE4 provides greater benefits compared with inhibiting either PDE alone. In respect to cAMP-dependent transcription, inhibitors of PDE3 (siguazodan, cilostazol) and PDE4 (rolipram, GSK256066, roflumilast N-oxide) each sensitized to the LABA, formoterol. This effect was magnified by dual PDE3 and PDE4 inhibition. Siguazodan plus rolipram was also more effective at inducing cAMP-dependent transcription than either inhibitor alone. Conversely, the concentration-response curve describing the enhancement of dexamethasone-induced, glucocorticoid response element-dependent transcription by formoterol was displaced to the left by PDE4, but not PDE3, inhibition. Overall, similar effects were described for bona fide genes, including RGS2, CD200, and CRISPLD2. Importantly, the combination of siguazodan plus rolipram prolonged the duration of gene expression induced by formoterol, dexamethasone, or dexamethasone plus formoterol. This was most apparent for RGS2, a bronchoprotective gene that may also reduce the proinflammatory effects of constrictor mediators. Collectively, these data provide a rationale for the use of PDE3 and PDE4 inhibitors in the treatment of COPD and asthma where they may enhance, sensitize, and prolong the effects of LABA/ICS combination therapies.
Collapse
Affiliation(s)
- Hawazen BinMahfouz
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bibhusana Borthakur
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dong Yan
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tresa George
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Newton
- Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
47
|
Zhang W, Zhi J, Cui Y, Zhang F, Habyarimana A, Cambier C, Gustin P. Potentiated interaction between ineffective doses of budesonide and formoterol to control the inhaled cadmium-induced up-regulation of metalloproteinases and acute pulmonary inflammation in rats. PLoS One 2014; 9:e109136. [PMID: 25313925 PMCID: PMC4196767 DOI: 10.1371/journal.pone.0109136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/28/2014] [Indexed: 02/02/2023] Open
Abstract
The anti-inflammatory properties of glucocorticoids are well known but their protective effects exerted with a low potency against heavy metals-induced pulmonary inflammation remain unclear. In this study, a model of acute pulmonary inflammation induced by a single inhalation of cadmium in male Sprague-Dawley rats was used to investigate whether formoterol can improve the anti-inflammatory effects of budesonide. The cadmium-related inflammatory responses, including matrix metalloproteinase-9 (MMP-9) activity, were evaluated. Compared to the values obtained in rats exposed to cadmium, pretreatment of inhaled budesonide (0.5 mg/15 ml) elicited a significant decrease in total cell and neutrophil counts in bronchoalveolar lavage fluid (BALF) associated with a significant reduction of MMP-9 activity which was highly correlated with the number of inflammatory cells in BALF. Additionally, cadmium-induced lung injuries characterized by inflammatory cell infiltration within alveoli and the interstitium were attenuated by the pre-treatment of budesonide. Though the low concentration of budesonide (0.25 mg/15 ml) exerted a very limited inhibitory effects in the present rat model, its combination with an inefficient concentration of formoterol (0.5 mg/30 ml) showed an enhanced inhibitory effect on neutrophil and total cell counts as well as on the histological lung injuries associated with a potentiation of inhibition on the MMP-9 activity. In conclusion, high concentration of budesonide alone could partially protect the lungs against cadmium exposure induced-acute neutrophilic pulmonary inflammation via the inhibition of MMP-9 activity. The combination with formoterol could enhance the protective effects of both drugs, suggesting a new therapeutic strategy for the treatment of heavy metals-induced lung diseases.
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- * E-mail:
| | - Jianming Zhi
- Department of Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongyao Cui
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fan Zhang
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Adélite Habyarimana
- Department for Functional Sciences B41, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Carole Cambier
- Department for Functional Sciences B41, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Pascal Gustin
- Department for Functional Sciences B41, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
48
|
Boardman C, Chachi L, Gavrila A, Keenan CR, Perry MM, Xia YC, Meurs H, Sharma P. Mechanisms of glucocorticoid action and insensitivity in airways disease. Pulm Pharmacol Ther 2014; 29:129-43. [PMID: 25218650 DOI: 10.1016/j.pupt.2014.08.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 01/04/2023]
Abstract
Glucocorticoids are the mainstay for the treatment of chronic inflammatory diseases including asthma and chronic obstructive pulmonary disease (COPD). However, it has been recognized that glucocorticoids do not work well in certain patient populations suggesting reduced sensitivity. The ultimate biologic responses to glucocorticoids are determined by not only the concentration of glucocorticoids but also the differences between individuals in glucocorticoid sensitivity, which is influenced by multiple factors. Studies are emerging to understand these mechanisms in detail, which would help in increasing glucocorticoid sensitivity in patients with chronic airways disease. This review aims to highlight both classical and emerging concepts of the anti-inflammatory mechanisms of glucocorticoids and also review some novel strategies to overcome steroid insensitivity in airways disease.
Collapse
Affiliation(s)
- C Boardman
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - L Chachi
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - A Gavrila
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - C R Keenan
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - M M Perry
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Y C Xia
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - H Meurs
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - P Sharma
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, 4C46 HRIC, 3280 Hospital Dr NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
49
|
Mikolka P, Mokrá D, Kopincová J, Tomčíková-Mikušiaková L, Calkovská A. Budesonide added to modified porcine surfactant Curosurf may additionally improve the lung functions in meconium aspiration syndrome. Physiol Res 2014; 62:S191-200. [PMID: 24329699 DOI: 10.33549/physiolres.932606] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Severe meconium aspiration syndrome (MAS) in newborns is often treated by exogenous surfactant. Because its efficacy is reduced by meconium-induced inflammation, glucocorticoid budesonide was added into surfactant preparation Curosurf to enhance efficacy of the surfactant therapy in experimental model of MAS. Oxygen-ventilated rabbits were intratracheally given meconium (25 mg/ml, 4 ml/kg) to induce respiratory failure. Thirty minutes later, animals were treated by intratracheal budesonide (0.25 mg/kg) or surfactant lung lavage (10 ml/kg, 5 mg phospholipids/ml) repeated twice, followed by undiluted Curosurf (100 mg phospholipids/kg) or by the above mentioned surfactant treatment with the last surfactant dose fortified with budesonide (0.25 mg/kg) or were untreated. Animals were ventilated for additional 5 hours and respiratory parameters were measured regularly. After sacrificing animals, wet-dry lung weight ratio was evaluated and plasma levels of interleukins (IL)-1beta, -6, -8, and TNF-alpha were measured by ELISA method. Efficacy of the given therapies to enhance lung functions and to diminish lung edema formation and inflammation increased from budesonide-only and surfactant-only therapy to surfactant+budesonide therapy. Combined therapy improved gas exchange from 30 min of administration, and showed a longer-lasting effect than surfactant-only therapy. In conclusions, budesonide additionally improved the effects of exogenous surfactant in experimental MAS.
Collapse
Affiliation(s)
- P Mikolka
- Department of Physiology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia.
| | | | | | | | | |
Collapse
|
50
|
Shi JX, Li JS, Hu R, Shi Y, Su X, Guo XJ, Li XM. Tristetraprolin is involved in the glucocorticoid-mediated interleukin 8 repression. Int Immunopharmacol 2014; 22:480-5. [PMID: 25111853 DOI: 10.1016/j.intimp.2014.07.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/20/2014] [Accepted: 07/27/2014] [Indexed: 11/29/2022]
Abstract
Glucocorticoids have been widely used in various inflammatory disorders, and the transcriptional repression of inflammatory mediators has been considered to be the main mechanism of action. However, a previous study showed that dexamethasone inhibited interleukin 8 (IL-8) expression by promoting IL-8 mRNA decay, which implies a posttranscriptional regulation. Nevertheless, by which mechanism dexamethasone destabilized IL-8 mRNA was unclear. Another study indicated that an RNA-binding protein, tristetraprolin (TTP), could be induced by dexamethasone. TTP can bind to AU-rich elements (ARE) in the 3'-untranslated region of target mRNAs and promotes mRNA degradation. So, we speculated that dexamethasone destabilized IL-8 mRNA by upregulating TTP expression. Here, we report that dexamethasone reduced IL-8 expression through destabilizing IL-8 mRNA in human pulmonary microvascular endothelial cells (HPMECs). Dexamethasone stimulation increased TTP mRNA and protein levels. TTP silencing led to mRNA stabilization and protein upregulation of IL-8. These results provide the evidence that the glucocorticoid, in HPMECs, inhibits IL-8 expression through TTP at the posttranscriptional level.
Collapse
Affiliation(s)
- Jia-Xin Shi
- Department of Respiratory Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Clinical Medical School of Nanjing Medical University, Lianyungang 222002, China.
| | - Jia-Shu Li
- Department of Respiratory Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Clinical Medical School of Nanjing Medical University, Lianyungang 222002, China.
| | - Rong Hu
- Department of Respiratory Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Clinical Medical School of Nanjing Medical University, Lianyungang 222002, China
| | - Yi Shi
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China.
| | - Xin Su
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Xiang-Jun Guo
- Department of Respiratory Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Clinical Medical School of Nanjing Medical University, Lianyungang 222002, China
| | - Xiao-Mei Li
- Department of Respiratory Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Clinical Medical School of Nanjing Medical University, Lianyungang 222002, China
| |
Collapse
|