1
|
Yue X, Peng L, Liu S, Zhang B, Zhang X, Chang H, Pei Y, Li X, Liu J, Shui W, Wu L, Xu H, Liu ZJ, Hua T. Structural basis of stepwise proton sensing-mediated GPCR activation. Cell Res 2025:10.1038/s41422-025-01092-w. [PMID: 40211064 DOI: 10.1038/s41422-025-01092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/23/2025] [Indexed: 04/12/2025] Open
Abstract
The regulation of pH homeostasis is crucial in many biological processes vital for survival, growth, and function of life. The pH-sensing G protein-coupled receptors (GPCRs), including GPR4, GPR65 and GPR68, play a pivotal role in detecting changes in extracellular proton concentrations, impacting both physiological and pathological states. However, comprehensive understanding of the proton sensing mechanism is still elusive. Here, we determined the cryo-electron microscopy structures of GPR4 and GPR65 in various activation states across different pH levels, coupled with Gs, Gq or G13 proteins, as well as a small molecule NE52-QQ57-bound inactive GPR4 structure. These structures reveal the dynamic nature of the extracellular loop 2 and its signature conformations in different receptor states, and disclose the proton sensing mechanism mediated by networks of extracellular histidine and carboxylic acid residues. Notably, we unexpectedly captured partially active intermediate states of both GPR4-Gs and GPR4-Gq complexes, and identified a unique allosteric binding site for NE52-QQ57 in GPR4. By integrating prior investigations with our structural analysis and mutagenesis data, we propose a detailed atomic model for stepwise proton sensation and GPCR activation. These insights may pave the way for the development of selective ligands and targeted therapeutic interventions for pH sensing-relevant diseases.
Collapse
Affiliation(s)
- Xiaolei Yue
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Li Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bingjie Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaodan Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hao Chang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuan Pei
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaoting Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Second Military Medical University, Shanghai, China.
- School of Clinical Medicine, Tsinghua University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
2
|
Wang H, Fung NH, Aloe C, Miles M, Selemidis S, Lopez AF, Wilson N, Owczarek C, Bozinovski S. CD131 antagonism blocks inflammation, emphysema and fibrosis in an asthma-COPD overlap mouse model originating in early life. Respirology 2025; 30:303-316. [PMID: 39814691 DOI: 10.1111/resp.14877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/15/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND AND OBJECTIVE Asthma-COPD overlap (ACO) is characterized by patients exhibiting features of both asthma and COPD. Currently, there is no specific treatment for ACO. This study aimed to investigate the therapeutic potential of targeting CD131, a shared receptor subunit for IL-3, IL-5 and GM-CSF, in ACO development and in preventing acute viral exacerbations. METHODS A two-hit mouse model of ACO was established by house dust mite (HDM) allergen sensitization to model asthma, and elastase treatment to model emphysema. In a separate model, human rhinovirus 1b (RV1b) was used to induce an acute asthma exacerbation. A neutralizing antibody against CD131 was used to block CD131 in vivo signalling. RESULTS Mice exposed to HDM and elastase developed cardinal features for asthma and COPD, including airway hyperreactivity (AHR) and emphysema. A mixed granulocytic inflammatory profile was identified in the lungs, including expansion of monocyte-derived macrophages, neutrophils and eosinophils. RT-qPCR analysis detected heightened gene expression of Mmp12, Il5 and Il13. Transcriptomic analysis further revealed pathway enrichment for type 2 inflammation and macrophage activation. Blockade of CD131 effectively reduced the lung inflammation and prevented the development of AHR, airway fibrosis and emphysema. Interestingly, pathway enrichment for Th1 response and interferon production detected in the model was not affected by the treatment. Consistently, CD131 antagonism prevented RV1b-induced asthma exacerbation without compromising RV1b clearance. CONCLUSION CD131 signalling coordinates multiple pathological pathways that drive airway inflammation and lung remodelling in ACO. Hence, CD131 antagonism represents a novel approach to combating the immunopathology in the complex ACO setting.
Collapse
Affiliation(s)
- Hao Wang
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Nok Him Fung
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Christian Aloe
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Mark Miles
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Angel F Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | | | | | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Matheson AM, Johnstone J, Niedbalski PJ, Woods JC, Castro M. New frontiers in asthma chest imaging. J Allergy Clin Immunol 2025; 155:241-254.e1. [PMID: 39709032 DOI: 10.1016/j.jaci.2024.12.1067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Modern pulmonary imaging can reveal underlying pathologic and pathophysiologic changes in the lungs of people with asthma, with important clinical implications. A multitude of imaging modalities, including computed tomography, magnetic resonance imaging, optical coherence tomography, and endobronchial ultrasound, are now being used to examine underlying structure-function relationships. Imaging-based biomarkers from these techniques, including airway dimensions, blood vessel volumes, mucus scores, extent of ventilation defect, and extent of air trapping, often have increased sensitivity compared with that of traditional lung function measurements and are increasingly being used as end points in clinical trials. Imaging has been crucial to recent improvements in our understanding of the relationships between type 2 inflammation, eosinophilia, and mucus extent. With the advent of effective anti-type 2 biologic therapies, computed tomography and magnetic resonance imaging techniques can identify not just which patients benefit from therapy but why they benefit. Clinical trials have begun to assess the utility of imaging to prospectively plan airway therapy targets in bronchial thermoplasty and have potential to direct future bronchoscopic therapies. Together, imaging techniques provide a diverse set of tools to investigate how spatially distributed airway, blood, and parenchymal abnormalities shape disease heterogeneity in patients with asthma.
Collapse
Affiliation(s)
- Alexander M Matheson
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph Johnstone
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Peter J Niedbalski
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan; Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, Kan
| | - Jason C Woods
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Cincinnati Bronchopulmonary Dysplasia Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mario Castro
- Pulmonary, Critical Care, and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kan.
| |
Collapse
|
4
|
Pattison LA, Rickman RH, Hilton H, Dannawi M, Wijesinghe SN, Ladds G, Yang LV, Jones SW, Smith ESJ. Activation of the proton-sensing GPCR, GPR65 on fibroblast-like synoviocytes contributes to inflammatory joint pain. Proc Natl Acad Sci U S A 2024; 121:e2410653121. [PMID: 39661058 PMCID: PMC11665855 DOI: 10.1073/pnas.2410653121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Inflammation is associated with localized acidosis, however, attributing physiological and pathological roles to proton-sensitive receptors is challenging due to their diversity and widespread expression. Here, agonists of the proton-sensing GPCR, GPR65, were systematically characterized. The synthetic agonist BTB09089 (BTB) recapitulated many proton-induced signaling events and demonstrated selectivity for GPR65. BTB was used to show that GPR65 activation on fibroblast-like synoviocytes (FLS), cells that line synovial joints, results in the secretion of proinflammatory mediators capable of recruiting immune cells and sensitizing sensory neurons. Intra-articular injection of BTB resulted in GPR65-dependent sensitization of knee-innervating neurons and nocifensive behaviors in mice. Stimulation of GPR65 on human FLS also triggered the release of inflammatory mediators and synovial fluid samples from human osteoarthritis patients were shown to activate GPR65. These results suggest a role of GPR65 in mediating cell-cell interactions that drive inflammatory joint pain in both mice and humans.
Collapse
Affiliation(s)
- Luke A. Pattison
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Maya Dannawi
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Li V. Yang
- Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC27834
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| |
Collapse
|
5
|
Liang J, Xia T, Wu S, Liu S, Guan Y. Application research on asthma-COPD overlap using low-dose CT scan and quantitative analysis. Clin Radiol 2024; 79:e1473-e1480. [PMID: 39384459 DOI: 10.1016/j.crad.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/23/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE The aim of this study was to assess the proximal airway remodeling, emphysema, and air trapping of asthma-COPD Overlap. MATERIALS AND METHODS 20 ACO patients, 55 mild to moderate COPD patients and 38 non-severe asthma patients were participated in low-dose dual phase CT scanning and pulmonary function test, comparative analysis was performed to identify differences in CT measurements among three groups. RESULTS (Ⅰ) The average age and smoking index of ACO and mild to moderate COPD were both higher than non-severe asthma. ACO and mild to moderate COPD group had a higher proportion of males than non-severe asthma. (Ⅱ) In terms of pulmonary function test, FEV1 (%Pred), FEV1/FVC%, MEF25% (%Pred), MEF 50% (%Pred), MMEF (%Pred), and PEF (%Pred) in ACO were significantly reduced than those in non-severe asthma. (Ⅲ) On proximal airway parameters, ACO exhibited higher WA% and Pi10 compared to mild to moderate COPD. However, there was no statistically significant difference in WA% and Pi10 between ACO and non-severe asthma. (Ⅳ) On CT lung function, the emphysema index VI-910ex of ACO was significantly higher than non-severe asthma.Additionally, ACO demonstrated higher MLDex and VI-856ex compared to non-severe asthma. CONCLUSIONS Compared with non-severe asthma, ACO is more common in male patients with older age and a longer history of smoking, which had more severe airflow obstruction and airway dysfunction than non-severe asthma.ACO showed more obvious proximal airway remodeling than mild to moderate COPD, and was more similar to non-severe asthma.The extent of emphysema and air trapping in ACO were more pronounced compared to non-severe asthma.
Collapse
Affiliation(s)
- J Liang
- Department of Radiology, Guangdong Provincial Second Hospital of Traditional Chinese Medicine, Guangzhou 510095, China
| | - T Xia
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - S Wu
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - S Liu
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Y Guan
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
| |
Collapse
|
6
|
Fangal VD, Saferali A, Castaldi PJ, Hersh CP, Weiss ST. Distinct physiological, transcriptomic, and imaging characteristics of asthma-COPD overlap compared to asthma and COPD in smokers. EBioMedicine 2024; 110:105453. [PMID: 39580967 PMCID: PMC11621799 DOI: 10.1016/j.ebiom.2024.105453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The clinical and pathological features of asthma and chronic obstructive pulmonary disease (COPD) can converge in smokers and elderly individuals as asthma-COPD overlap (ACO). This overlap challenges the diagnosis and treatment of the distinct aetiologies underlying these conditions. METHODS We analysed 2453 smokers (≥10 pack-years), aged 45-80 years, from the Genetic Epidemiology of COPD (COPDGene) Study, stratified as Control, Asthma, COPD, and ACO based on Global Initiative for Chronic Obstructive Lung Disease (GOLD) criteria. A comprehensive assessment was performed, encompassing symptomatology, pulmonary function tests (PFTs), complete blood counts (CBCs), bulk RNA sequencing (RNA-seq), and high-resolution quantitative computed tomography (QCT) imaging to evaluate clinical impact, lung function, systemic inflammation, and structural alterations contributing to disease progression across respiratory phenotypes. Differential expression (DE) analysis was performed using whole blood RNA-seq (BH-corrected FDR < 0.01), followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Group differences were assessed using the Mann-Whitney U-test (MWU) or Chi-squared test (χ2), with Bonferroni correction applied for multiple comparisons. Multivariate linear regression models were used to adjust the associations between disease status and specific clinical outcomes for confounders, with one-way ANOVA and Tukey's Honest Significant Difference (HSD) post-hoc test applied for pairwise comparisons. Our analysis aimed to delineate the extent and variability of clinical features among disease phenotypes to guide targeted therapeutic strategies. FINDINGS Our study highlights distinct yet overlapping profiles across ACO, asthma, and COPD. We effectively isolated disease-specific mechanisms by comparing each phenotype to smoking controls (GOLD 0) while accounting for baseline smoking-related inflammation. ACO exhibited the most severe symptom burden, with significantly higher COPD Assessment Test (CAT) score (18.32, 95% CI: [17.02, 19.63], P < 0.0001) and Modified Medical Research Council (mMRC) Dyspnea score (2.14, 95% CI: [1.92, 2.35], P < 0.0001) compared to COPD and asthma. ACO also displayed reduced lung capacity (forced expiratory volume in 1 s [FEV1]: 52.5%, 95% CI: [50.08, 54.93], P < 0.0001) and airflow limitation (FEV1/forced vital capacity [FVC]: 0.55, 95% CI: [0.5471, 0.5546], P < 0.0001), closely resembling COPD but significantly worse than asthma. The inflammatory profile of ACO exhibited a mixed response, featuring elevated neutrophil counts (4.57 K/μL, 95% CI: [4.28, 4.86], P < 0.0001) and eosinophil levels (0.22 K/μL, 95% CI: [0.20, 0.25], P < 0.01), contrasting with the predominantly neutrophilic inflammation in COPD and the absence of systemic inflammation in asthma. Structurally, ACO demonstrated significant airway remodelling (Pi10: 2.87, 95% CI: [2.83, 2.91], P < 0.0001), intermediate emphysema (5.66%, 95% CI: [4.72, 6.60], P < 0.0001), and moderate small airway disease (parametric response mapping for functional small airway disease [PRMfSAD]: 22.94%, 95% CI: [21.53, 24.34], P < 0.0001), reflecting features of both asthma and COPD. COPD was characterised by more extensive emphysema (8.9%, 95% CI: [8.34, 9.45], P < 0.0001), small airway disease (PRMfSAD: 27.09%, 95% CI: [26.51, 27.66], P < 0.0001), and gas trapping (37.34%, 95% CI: [36.33, 38.35], P < 0.0001), alongside moderate airway remodelling. At a molecular level, DE analysis revealed enrichment of the Hypoxia-Inducible Factor 1 (HIF-1) pathway in ACO, highlighting unique hypoxia-driven metabolic adaptations, while COPD was associated with neutrophil extracellular trap (NET) formation and necroptosis. In contrast, asthma exhibited significant airway remodelling (Pi10: 2.09, 95% CI: [2.05, 2.13], P < 0.0001), minimal parenchymal damage, and no systemic gene expression changes. INTERPRETATION Collectively, our findings underscore the lung function impairments, systemic inflammation, molecular mechanisms, and structural correlates distinguishing ACO from COPD and asthma, emphasising the need for precise clinical management and the potential for novel therapeutic interventions. FUNDING This work was supported by National Heart, Lung, and Blood Institute (NHLBI) grants U01 HL089897 and U01 HL089856, as well as by National Institutes of Health (NIH) contract 75N92023D00011. Additional support was provided by grants R01 HL166231 (C.P.H.) and K01 HL157613 (A.S.).
Collapse
Affiliation(s)
- Vrushali D Fangal
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
7
|
Aljama C, Granados G, Callejas-González FJ, Martínez-Rivera C, Pallarés-Sanmartín A, Rodríguez-Pons L, Cabrera-César E, Márquez-Martín E, Boldova-Loscertales A, Naval-Sendra E, Abascal-Bolado B, Cabrera-López C, Miravitlles M, Esquinas C, Barrecheguren M. Comparison of the Determinants of the "Chronic Obstructive Pulmonary Disease Assessment Test" (CAT) and the "Asthma Control Test" (ACT) in Patients with Asthma-COPD Overlap. J Clin Med 2024; 13:6367. [PMID: 39518506 PMCID: PMC11546969 DOI: 10.3390/jcm13216367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Objective: The objective of this study was to investigate which of two short questionnaires, the Asthma Control Test (ACT) or the COPD Assessment Test (CAT), correlates better with severity variables and whether they share similar determinants in patients with asthma-COPD overlap. Method: This observational, cross-sectional, multicentric study included smokers and former smokers of more than 10 pack-years, with non-fully reversible airflow obstruction and either a concomitant diagnosis of asthma or signs of type 2 inflammation, from 15 centres in Spain. Results: A total of 157 patients were included, 109 (69.4%) were men, the mean age was 63.3 (SD: 9) years and the mean FEV1 (%) was 59.7% (SD: 20.5%). The mean CAT score was 14.5 (SD: 8.7), and the mean ACT score was 17.9 (SD: 5.2). Both scores showed good correlations (r = 0.717; p < 0.001). In the multivariate analysis, the Hospital Anxiety and Depression Scale and mMRC dyspnoea scores were independently and significantly associated with both the CAT and ACT scores; however, age was only significantly associated with the CAT, and the EQ-5D scores and the number of exacerbations in the previous year were only significantly associated with the ACT scores. The ACT had a slightly better predictive value for exacerbations than the CAT (AUC = 0.70 (95% CI: 0.62 to 0.79 vs. 0.65 (95% CI: 0.56 to 0.74))). Conclusions: There is a good correlation between ACT and CAT scores in patients with ACO. However, severe patients scored worse on the CAT than the ACT. Anxiety, depression and dyspnoea were significantly associated with both the CAT and ACT scores. The ACT was a slightly better predictor of exacerbations than the CAT in this population.
Collapse
Affiliation(s)
- Cristina Aljama
- Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (G.G.); (C.E.); (M.B.)
| | - Galo Granados
- Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (G.G.); (C.E.); (M.B.)
| | | | - Carlos Martínez-Rivera
- Pneumology Department, Hospital Germans Trias i Pujol, 08916 Badalona, Spain; (C.M.-R.); (L.R.-P.)
| | - Abel Pallarés-Sanmartín
- Pneumology Department, Complejo Hospitalario Universitario de Ourense, 32005 Ourense, Spain;
| | - Laura Rodríguez-Pons
- Pneumology Department, Hospital Germans Trias i Pujol, 08916 Badalona, Spain; (C.M.-R.); (L.R.-P.)
| | - Eva Cabrera-César
- Pneumology Department, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain;
| | | | | | - Elsa Naval-Sendra
- Pneumology Department, Hospital Universitario de La Ribera, 46600 Alzira, Spain;
| | - Beatriz Abascal-Bolado
- Pneumology Department, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain;
| | - Carlos Cabrera-López
- Pneumology Department, Hospital Universitario Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain;
| | - Marc Miravitlles
- Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (G.G.); (C.E.); (M.B.)
| | - Cristina Esquinas
- Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (G.G.); (C.E.); (M.B.)
| | - Miriam Barrecheguren
- Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (G.G.); (C.E.); (M.B.)
| |
Collapse
|
8
|
Morris SB, Ocadiz-Ruiz R, Asai N, Malinczak CA, Rasky AJ, Lombardo GK, Velarde EM, Ptaschinski C, Zemans RL, Lukacs NW, Fonseca W. Long-term alterations in lung epithelial cells after EL-RSV infection exacerbate allergic responses through IL-1β-induced pathways. Mucosal Immunol 2024; 17:1072-1088. [PMID: 39069078 PMCID: PMC11610113 DOI: 10.1016/j.mucimm.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
Early-life (EL) respiratory infections increase pulmonary disease risk, especially EL-Respiratory Syncytial Virus (EL-RSV) infections linked to asthma. Mechanisms underlying asthma predisposition remain unknown. In this study, we examined the long-term effects on the lung after four weeks post EL-RSV infection. We identified alterations in the lung epithelial cell, with a rise in the percentage of alveolar type 2 epithelial cells (AT2) and a decreased percentage of cells in the AT1 and AT2-AT1 subclusters, as well as upregulation of Bmp2 and Krt8 genes that are associated with AT2-AT1 trans-differentiation, suggesting potential defects in lung repair processes. We identified persistent upregulation of asthma-associated genes, including Il33. EL-RSV-infected mice allergen-challenged exhibited exacerbated allergic response, with significant upregulation of Il33 in the lung and AT2 cells. Similar long-term effects were observed in mice exposed to EL-IL-1β. Notably, treatment with IL-1ra during acute EL-RSV infection mitigated the long-term alveolar alterations and the allergen-exacerbated response. Finally, epigenetic modifications in the promoter of the Il33 gene were detected in AT2 cells harvested from EL-RSV and EL-IL1β groups, suggesting that long-term alteration in the epithelium after RSV infection is dependent on the IL-1β pathway. This study provides insight into the molecular mechanisms of asthma predisposition after RSV infection.
Collapse
Affiliation(s)
- Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ramon Ocadiz-Ruiz
- Department of Bioengineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nobuhiro Asai
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace K Lombardo
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan M Velarde
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L Zemans
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
9
|
Justus CR, Marie MA, Sanderlin EJ, Yang LV. The Roles of Proton-Sensing G-Protein-Coupled Receptors in Inflammation and Cancer. Genes (Basel) 2024; 15:1151. [PMID: 39336742 PMCID: PMC11431078 DOI: 10.3390/genes15091151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The precise regulation of pH homeostasis is crucial for normal physiology. However, in tissue microenvironments, it can be impacted by pathological conditions such as inflammation and cancer. Due to the overproduction and accumulation of acids (protons), the extracellular pH is characteristically more acidic in inflamed tissues and tumors in comparison to normal tissues. A family of proton-sensing G-protein-coupled receptors (GPCRs) has been identified as molecular sensors for cells responding to acidic tissue microenvironments. Herein, we review the current research progress pertaining to these proton-sensing GPCRs, including GPR4, GPR65 (TDAG8), and GPR68 (OGR1), in inflammation and cancer. Growing evidence suggests that GPR4 and GPR68 are mainly pro-inflammatory, whereas GPR65 is primarily anti-inflammatory, in various inflammatory disorders. Both anti- and pro-tumorigenic effects have been reported for this family of receptors. Moreover, antagonists and agonists targeting proton-sensing GPCRs have been developed and evaluated in preclinical models. Further research is warranted to better understand the roles of these proton-sensing GPCRs in pathophysiology and is required in order to exploit them as potential therapeutic targets for disease treatment.
Collapse
Affiliation(s)
- Calvin R Justus
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
10
|
Sayers I, John C, Chen J, Hall IP. Genetics of chronic respiratory disease. Nat Rev Genet 2024; 25:534-547. [PMID: 38448562 DOI: 10.1038/s41576-024-00695-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 03/08/2024]
Abstract
Chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), asthma and interstitial lung diseases are frequently occurring disorders with a polygenic basis that account for a large global burden of morbidity and mortality. Recent large-scale genetic epidemiology studies have identified associations between genetic variation and individual respiratory diseases and linked specific genetic variants to quantitative traits related to lung function. These associations have improved our understanding of the genetic basis and mechanisms underlying common lung diseases. Moreover, examining the overlap between genetic associations of different respiratory conditions, along with evidence for gene-environment interactions, has yielded additional biological insights into affected molecular pathways. This genetic information could inform the assessment of respiratory disease risk and contribute to stratified treatment approaches.
Collapse
Affiliation(s)
- Ian Sayers
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, University Park, Nottingham, UK
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, UK
| | - Catherine John
- University of Leicester, Leicester, UK
- University Hospitals of Leicester, Leicester, UK
| | - Jing Chen
- University of Leicester, Leicester, UK
| | - Ian P Hall
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, University Park, Nottingham, UK.
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, UK.
| |
Collapse
|
11
|
Tanabe N, Matsumoto H, Morimoto C, Hayashi Y, Sakamoto R, Oguma T, Nagasaki T, Sunadome H, Sato A, Sato S, Ohashi K, Tsukahara T, Hirai T. Mucus plugging on computed tomography and the sputum microbiome in patients with asthma, chronic obstructive pulmonary disease, and asthma-COPD overlap. Allergol Int 2024:S1323-8930(24)00055-8. [PMID: 39013753 DOI: 10.1016/j.alit.2024.05.004] [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: 03/13/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Despite clinical implications, the pathogenesis of mucus plugging in asthma, chronic obstructive pulmonary disease (COPD), and asthma-COPD overlap (ACO) remains unclear. We hypothesized that distinct airway microbiomes might affect mucus plugging differently among ACO, asthma, and COPD and among different extents of airway eosinophilic inflammation. METHODS The sputum microbiome, sputum cell differential count, and mucus plug score on computed tomography were cross-sectionally evaluated in patients with chronic airflow limitation. RESULTS Patients with ACO, asthma, or COPD were enrolled (n = 56, 10, and 25). Higher mucus plug scores were associated with a greater relative abundance of the phylum Proteobacteria (rho = 0.29) only in patients with ACO and a greater relative abundance of the phylum Actinobacteria (rho = 0.46) only in patients with COPD. In multivariable models including only patients with ACO, the presence of mucus plugs was associated with a greater relative abundance of the phylum Proteobacteria and the genus Haemophilus, independent of smoking status, airflow limitation, and emphysema severity. Moreover, the mucus score was associated with a greater relative abundance of the genus Streptococcus (rho = 0.46) in patients with a high sputum eosinophil count (n = 22) and with that of the genus Haemophilus (rho = 0.46) in those with a moderate sputum eosinophil count (n = 26). CONCLUSIONS The associations between mucus plugging and the microbiome in ACO differed from those in COPD and asthma. Greater relative abundances of the phylum Proteobacteria and genus Haemophilus may be involved in mucus plugging in patients with ACO and moderate airway eosinophilic inflammation.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Medicine & Allergology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Chie Morimoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yusuke Hayashi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryo Sakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadao Nagasaki
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hironobu Sunadome
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsuyasu Sato
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kai Ohashi
- Kyoto Institute of Nutrition & Pathology, Inc., Kyoto, Japan
| | | | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Chen Z, Shang Y, Zhang X, Duan W, Li J, Zhu L, Ma L, Xiang X, Jia J, Ji X, Gong S. METTL3 mediates SOX5 m6A methylation in bronchial epithelial cells to attenuate Th2 cell differentiation in T2 asthma. Heliyon 2024; 10:e28884. [PMID: 38601672 PMCID: PMC11004579 DOI: 10.1016/j.heliyon.2024.e28884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Asthma, a chronic inflammatory disease in which type 2 T helper cells (Th2) play a causative role in the development of T2 asthma. N6-methyladenosine (m6A) modification, an mRNA modification, and methyltransferase-like 3 (METTL3) is involved in the development of T2 asthma by inhibiting Th2 cell differentiation. Sex determining region Y-box protein 5 (SOX5) is involved in regulating T cell differentiation, but its role in T2 asthma was unclear. The objective of this study was to explore the role of METTL3 and SOX5 in T2 asthma and whether there is an interaction between the two. Materials and methods Adults diagnosed with T2 asthma (n = 14) underwent clinical information collection and pulmonary function tests. In vivo and in vitro T2 asthma models were established using female C57BL/6 mice and human bronchial epithelial cells (HBE). The expressions of METTL3 and SOX5 were detected by Western blot and qRT-PCR and Western blot. Th2 cell differentiation was determined by flow cytometry and IL-4 level was detected by ELISA. m6A methylation level was determined by m6A quantitative assay. The relationship between METTL3 expression and clinical parameters was determined by Spearman rank correlation analysis. The function of METTL3 and SOX5 genes in asthma was investigated in vitro and in vivo. The RNA immunoprecipitation assay detected the specific interaction between METTL3 and SOX5. Results Patients with T2 asthma displayed lower METTL3 levels compared to healthy controls. Within this group, a negative correlation was observed between METTL3 and Th2 cells, while a positive correlation was noted between METTL3 and clinical parameters as well as Th1 cells. In both in vitro and in vivo models representing T2 asthma, METTL3 levels decreased significantly, while SOX5 levels showed the opposite trend. Overexpression of METTL3 gene in HBE cells significantly inhibited Th2 cell differentiation and increased m6A methylation activity. From a mechanism perspective, low METTL3 negatively regulates SOX5 expression through m6A modification dependence, while high SOX5 expression is positively associated with T2 asthma severity. Cell transfection experiments confirmed that METTL3 regulates Th2 cell differentiation and IL-4 release through SOX5. Conclusions Overall, our results indicate that METTL3 alleviates Th2 cell differentiation in T2 asthma by modulating the m6A methylation activity of SOX5 in bronchial epithelial cells. This mechanism could potentially serve as a target for the prevention and management of T2 asthma.
Collapse
Affiliation(s)
- Zhifeng Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yulin Shang
- Ophthalmology and Otorhinolaryngology, Zigui County Traditional Chinese Medicine Hospital, 30 Pinghu Avenue, Zigui, Hubei, 443600, China
| | - Xiufeng Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Hainan Medical University, 48 Pak Shui Tong Road, Haikou, Hainan, 570000, China
| | - Wentao Duan
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital, 61 West Jiefang Road, Changsha, Hunan, 410005, China
| | - Jianmin Li
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital, 61 West Jiefang Road, Changsha, Hunan, 410005, China
| | - Liming Zhu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital, 61 West Jiefang Road, Changsha, Hunan, 410005, China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University, 15 Le Qun Road, Guilin, Guangxi, 541001, China
| | - Xudong Xiang
- Department of Emergency, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Jingsi Jia
- Department of Emergency, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Xiaoying Ji
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Guiyang, Guizhou, 550004, China
| | - Subo Gong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| |
Collapse
|
13
|
Hausmann M, Seuwen K, de Vallière C, Busch M, Ruiz PA, Rogler G. Role of pH-sensing receptors in colitis. Pflugers Arch 2024; 476:611-622. [PMID: 38514581 PMCID: PMC11006753 DOI: 10.1007/s00424-024-02943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis-two contributing factors to disease pathophysiology-are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Collapse
Affiliation(s)
- Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland.
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Moana Busch
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| |
Collapse
|
14
|
Yang W, Li Z, Yang T, Li Y, Xie Z, Feng L, Peng Z, Liu J. Experts' Consensus on the Management of Respiratory Disease Syndemic. China CDC Wkly 2024; 6:131-138. [PMID: 38476822 PMCID: PMC10926044 DOI: 10.46234/ccdcw2024.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Affiliation(s)
- Weizhong Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control, Peking Union Medical College, Ministry of Education, Beijing, China
| | - Zhongjie Li
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control, Peking Union Medical College, Ministry of Education, Beijing, China
| | - Ting Yang
- National Center for Respiratory Diseases, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yanming Li
- Department of Pulmonary and Critical Care Medicine, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhengde Xie
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, 2019RU016, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control, Peking Union Medical College, Ministry of Education, Beijing, China
| | - Zhibin Peng
- Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of PublicHealth, Key Laboratory of Epidemiology of Major Diseases, Ministry of Education, Peking University Health Science Center-Weifang Joint Research Center for Maternal and Child Health,Peking University, Beijing, China
| |
Collapse
|
15
|
Edris A, Voorhies K, Lutz SM, Iribarren C, Hall I, Wu AC, Tobin M, Fawcett K, Lahousse L. Asthma exacerbations and eosinophilia in the UK Biobank: a genome-wide association study. ERJ Open Res 2024; 10:00566-2023. [PMID: 38196893 PMCID: PMC10772900 DOI: 10.1183/23120541.00566-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/31/2023] [Indexed: 01/11/2024] Open
Abstract
Background Asthma exacerbations reflect disease severity, affect morbidity and mortality, and may lead to declining lung function. Inflammatory endotypes (e.g. T2-high (eosinophilic)) may play a key role in asthma exacerbations. We aimed to assess whether genetic susceptibility underlies asthma exacerbation risk and additionally tested for an interaction between genetic variants and eosinophilia on exacerbation risk. Methods UK Biobank data were used to perform a genome-wide association study of individuals with asthma and at least one exacerbation compared to individuals with asthma and no history of exacerbations. Individuals with asthma were identified using self-reported data, hospitalisation data and general practitioner records. Exacerbations were identified as either asthma-related hospitalisation, general practitioner record of asthma exacerbation or an oral corticosteroid burst prescription. A logistic regression model adjusted for age, sex, smoking status and genetic ancestry via principal components was used to assess the association between genetic variants and asthma exacerbations. We sought replication for suggestive associations (p<5×10-6) in the GERA cohort. Results In the UK Biobank, we identified 11 604 cases and 37 890 controls. While no variants reached genome-wide significance (p<5×10-8) in the primary analysis, 116 signals were suggestively significant (p<5×10-6). In GERA, two single nucleotide polymorphisms (rs34643691 and rs149721630) replicated (p<0.05), representing signals near the NTRK3 and ABCA13 genes. Conclusions Our study has identified reproducible associations with asthma exacerbations in the UK Biobank and GERA cohorts. Confirmation of these findings in different asthma subphenotypes in diverse ancestries and functional investigation will be required to understand their mechanisms of action and potentially inform therapeutic development.
Collapse
Affiliation(s)
- Ahmed Edris
- Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Kirsten Voorhies
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Sharon M. Lutz
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Ian Hall
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Ann Chen Wu
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Martin Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Katherine Fawcett
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
- These authors contributed equally
| | - Lies Lahousse
- Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- These authors contributed equally
| |
Collapse
|
16
|
Odimba U, Senthilselvan A, Farrell J, Gao Z. Sex-Specific Genetic Determinants of Asthma-COPD Phenotype and COPD in Middle-Aged and Older Canadian Adults: An Analysis of CLSA Data. COPD 2023; 20:233-247. [PMID: 37466093 DOI: 10.1080/15412555.2023.2229906] [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: 02/17/2023] [Revised: 05/22/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
The etiology of sex differences in the risk of asthma-COPD phenotype and COPD is still not completely understood. Genetic and environmental risk factors are commonly believed to play an important role. This study aims to identify sex-specific genetic markers associated with asthma-COPD phenotype and COPD using the Canadian Longitudinal Study on Aging (CLSA) Baseline Comprehensive and Genomic data. There were a total of 1,415 COPD cases. Out of them, 504 asthma-COPD phenotype cases were identified. 20,524 participants without a diagnosis of asthma and COPD served as controls. We performed genome-wide SNP-by-sex interaction analysis. SNPs with an interaction p-value < 10-5 were included in a sex-stratified multivariable logistic regression for asthma-COPD phenotype and COPD outcomes. 18 and 28 SNPs had a significant interaction term p-value < 10-5 with sex in the regression analyses of asthma-COPD phenotype and COPD outcomes, respectively. Sex-stratified multivariable analysis of asthma-COPD phenotype showed that 7 SNPs in/near SMYD3, FHIT, ZNF608, RIMBP2, ZNF133, BPIFB1, and S100B loci were significant in males. Sex-stratified multivariable analysis of COPD showed that 8 SNPs in/near MAGI1, COX18, OSTC, ELOVL5, C7orf72 FGF14, and NKAIN4 were significant in males, and 4 SNPs in/near genes CAMTA1, SATB2, PDE10A, and LINC00908 were significant in females. An SNP in the ZPBP gene was associated with COPD in both males and females. Identification of sex-specific loci associated with asthma-COPD phenotype and COPD may offer valuable evidence toward a better understanding of the sex-specific differences in the pathophysiology of the diseases.
Collapse
Affiliation(s)
- Ugochukwu Odimba
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
| | | | - Jamie Farrell
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
- Faculty of Medicine, Health Sciences Centre (Respirology Department), Memorial University, St John's, Newfoundland and Labrador, Canada
| | - Zhiwei Gao
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
| |
Collapse
|
17
|
Moll M, Sordillo JE, Ghosh AJ, Hayden LP, McDermott G, McGeachie MJ, Dahlin A, Tiwari A, Manmadkar MG, Abston ED, Pavuluri C, Saferali A, Begum S, Ziniti JP, Gulsvik A, Bakke PS, Aschard H, Iribarren C, Hersh CP, Sparks JA, Hobbs BD, Lasky-Su JA, Silverman EK, Weiss ST, Wu AC, Cho MH. Polygenic risk scores identify heterogeneity in asthma and chronic obstructive pulmonary disease. J Allergy Clin Immunol 2023; 152:1423-1432. [PMID: 37595761 PMCID: PMC10841234 DOI: 10.1016/j.jaci.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Asthma and chronic obstructive pulmonary disease (COPD) have distinct and overlapping genetic and clinical features. OBJECTIVE We sought to test the hypothesis that polygenic risk scores (PRSs) for asthma (PRSAsthma) and spirometry (FEV1 and FEV1/forced vital capacity; PRSspiro) would demonstrate differential associations with asthma, COPD, and asthma-COPD overlap (ACO). METHODS We developed and tested 2 asthma PRSs and applied the higher performing PRSAsthma and a previously published PRSspiro to research (Genetic Epidemiology of COPD study and Childhood Asthma Management Program, with spirometry) and electronic health record-based (Mass General Brigham Biobank and Genetic Epidemiology Research on Adult Health and Aging [GERA]) studies. We assessed the association of PRSs with COPD and asthma using modified random-effects and binary-effects meta-analyses, and ACO and asthma exacerbations in specific cohorts. Models were adjusted for confounders and genetic ancestry. RESULTS In meta-analyses of 102,477 participants, the PRSAsthma (odds ratio [OR] per SD, 1.16 [95% CI, 1.14-1.19]) and PRSspiro (OR per SD, 1.19 [95% CI, 1.17-1.22]) both predicted asthma, whereas the PRSspiro predicted COPD (OR per SD, 1.25 [95% CI, 1.21-1.30]). However, results differed by cohort. The PRSspiro was not associated with COPD in GERA and Mass General Brigham Biobank. In the Genetic Epidemiology of COPD study, the PRSAsthma (OR per SD: Whites, 1.3; African Americans, 1.2) and PRSspiro (OR per SD: Whites, 2.2; African Americans, 1.6) were both associated with ACO. In GERA, the PRSAsthma was associated with asthma exacerbations (OR, 1.18) in Whites; the PRSspiro was associated with asthma exacerbations in White, LatinX, and East Asian participants. CONCLUSIONS PRSs for asthma and spirometry are both associated with ACO and asthma exacerbations. Genetic prediction performance differs in research versus electronic health record-based cohorts.
Collapse
Affiliation(s)
- Matthew Moll
- Department of Medicine, Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Mass; Harvard Medical School, Brigham and Women's Hospital, Boston, Mass
| | - Joanne E Sordillo
- Department of Population Medicine, PRecisiOn Medicine Translational Research (PROMoTeR) Center, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Mass
| | - Auyon J Ghosh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, SUNY Upstate Medical Center, Syracuse, NY
| | - Lystra P Hayden
- Department of Pediatrics, Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Massachusetts General Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Gregory McDermott
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Mass
| | - Michael J McGeachie
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Amber Dahlin
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Anshul Tiwari
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Monica G Manmadkar
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Eric D Abston
- Department of Thoracic Surgery, Massachusetts General Hospital, Boston, Mass
| | - Chandan Pavuluri
- Department of Medicine, Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Mass; Harvard Medical School, Brigham and Women's Hospital, Boston, Mass
| | - Aabida Saferali
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Sofina Begum
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - John P Ziniti
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Per S Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hugues Aschard
- Department of Computational Biology, Institut Pasteur, Universit de Paris, Paris, France
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, Calif
| | - Craig P Hersh
- Department of Medicine, Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Mass; Harvard Medical School, Brigham and Women's Hospital, Boston, Mass
| | - Jeffrey A Sparks
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Mass
| | - Brian D Hobbs
- Department of Medicine, Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Mass; Harvard Medical School, Brigham and Women's Hospital, Boston, Mass
| | - Jessica A Lasky-Su
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Edwin K Silverman
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Scott T Weiss
- Harvard Medical School, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, Mass
| | - Ann Chen Wu
- Department of Population Medicine, PRecisiOn Medicine Translational Research (PROMoTeR) Center, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Mass
| | - Michael H Cho
- Department of Medicine, Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Mass; Harvard Medical School, Brigham and Women's Hospital, Boston, Mass.
| |
Collapse
|
18
|
Perren L, Busch M, Schuler C, Ruiz PA, Foti F, Weibel N, de Vallière C, Morsy Y, Seuwen K, Hausmann M, Rogler G. OGR1 (GPR68) and TDAG8 (GPR65) Have Antagonistic Effects in Models of Colonic Inflammation. Int J Mol Sci 2023; 24:14855. [PMID: 37834303 PMCID: PMC10573511 DOI: 10.3390/ijms241914855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
G-protein-coupled receptors (GPRs), including pro-inflammatory ovarian cancer GPR1 (OGR1/GPR68) and anti-inflammatory T cell death-associated gene 8 (TDAG8/GPR65), are involved in pH sensing and linked to inflammatory bowel disease (IBD). OGR1 and TDAG8 show opposite effects. To determine which effect is predominant or physiologically more relevant, we deleted both receptors in models of intestinal inflammation. Combined Ogr1 and Tdag8 deficiency was assessed in spontaneous and acute murine colitis models. Disease severity was assessed using clinical scores. Colon samples were analyzed using quantitative polymerase chain reaction (qPCR) and flow cytometry (FACS). In acute colitis, Ogr1-deficient mice showed significantly decreased clinical scores compared with wildtype (WT) mice, while Tdag8-deficient mice and double knockout (KO) mice presented similar scores to WT. In Il-10-spontaneous colitis, Ogr1-deficient mice presented significantly decreased, and Tdag8-deficient mice had increased inflammation. In the Il10-/- × Ogr1-/- × Tdag8-/- triple KO mice, inflammation was significantly decreased compared with Tdag8-/-. Absence of Ogr1 reduced pro-inflammatory cytokines in Tdag8-deficient mice. Tdag8-/- had significantly more IFNγ+ T-lymphocytes and IL-23 T-helper cells in the colon compared with WT. The absence of OGR1 significantly alleviates the intestinal damage mediated by the lack of functional TDAG8. Both OGR1 and TDAG8 represent potential new targets for therapeutic intervention.
Collapse
|
19
|
Plaza Moral V, Alobid I, Álvarez Rodríguez C, Blanco Aparicio M, Ferreira J, García G, Gómez-Outes A, Garín Escrivá N, Gómez Ruiz F, Hidalgo Requena A, Korta Murua J, Molina París J, Pellegrini Belinchón FJ, Plaza Zamora J, Praena Crespo M, Quirce Gancedo S, Sanz Ortega J, Soto Campos JG. GEMA 5.3. Spanish Guideline on the Management of Asthma. OPEN RESPIRATORY ARCHIVES 2023; 5:100277. [PMID: 37886027 PMCID: PMC10598226 DOI: 10.1016/j.opresp.2023.100277] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
The Spanish Guideline on the Management of Asthma, better known by its acronym in Spanish GEMA, has been available for more than 20 years. Twenty-one scientific societies or related groups both from Spain and internationally have participated in the preparation and development of the updated edition of GEMA, which in fact has been currently positioned as the reference guide on asthma in the Spanish language worldwide. Its objective is to prevent and improve the clinical situation of people with asthma by increasing the knowledge of healthcare professionals involved in their care. Its purpose is to convert scientific evidence into simple and easy-to-follow practical recommendations. Therefore, it is not a monograph that brings together all the scientific knowledge about the disease, but rather a brief document with the essentials, designed to be applied quickly in routine clinical practice. The guidelines are necessarily multidisciplinary, developed to be useful and an indispensable tool for physicians of different specialties, as well as nurses and pharmacists. Probably the most outstanding aspects of the guide are the recommendations to: establish the diagnosis of asthma using a sequential algorithm based on objective diagnostic tests; the follow-up of patients, preferably based on the strategy of achieving and maintaining control of the disease; treatment according to the level of severity of asthma, using six steps from least to greatest need of pharmaceutical drugs, and the treatment algorithm for the indication of biologics in patients with severe uncontrolled asthma based on phenotypes. And now, in addition to that, there is a novelty for easy use and follow-up through a computer application based on the chatbot-type conversational artificial intelligence (ia-GEMA).
Collapse
Affiliation(s)
| | - Isam Alobid
- Otorrinolaringología, Hospital Clinic de Barcelona, España
| | | | | | - Jorge Ferreira
- Hospital de São Sebastião – CHEDV, Santa Maria da Feira, Portugal
| | | | - Antonio Gómez-Outes
- Farmacología clínica, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), Madrid, España
| | - Noé Garín Escrivá
- Farmacia Hospitalaria, Hospital de la Santa Creu i Sant Pau, Barcelona, España
| | | | | | - Javier Korta Murua
- Neumología Pediátrica, Hospital Universitario Donostia, Donostia-San, Sebastián, España
| | - Jesús Molina París
- Medicina de familia, semFYC, Centro de Salud Francia, Fuenlabrada, Dirección Asistencial Oeste, Madrid, España
| | | | - Javier Plaza Zamora
- Farmacia comunitaria, Farmacia Dr, Javier Plaza Zamora, Mazarrón, Murcia, España
| | | | | | - José Sanz Ortega
- Alergología Pediátrica, Hospital Católico Universitario Casa de Salud, Valencia, España
| | | |
Collapse
|
20
|
Peng K, Li Z, Gao TR, Lv J, Wang WJ, Zhan P, Yao WC, Zhao H, Wang H, Xu DX, Huang Y, Tan ZX. Polycyclic aromatic hydrocarbon exposure burden: Individual and mixture analyses of associations with chronic obstructive pulmonary disease risk. ENVIRONMENTAL RESEARCH 2023; 222:115334. [PMID: 36702192 DOI: 10.1016/j.envres.2023.115334] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/01/2022] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
Accumulating data demonstrate that polycyclic aromatic hydrocarbons (PAH) exposure is linked to compromised respiratory diseases. This study aimed to analyze urinary PAH metabolites and their associations with chronic obstructive pulmonary disease (COPD) in a sample size of 3015 subjects from a total population of 50,588 from the National Health and Nutrition Examination Survey (NHANES) in 2007-2016. Results showed that the most predominant metabolite was 1-Hydroxynaphthalene (1-NAP, 84%) with a geometric mean concentration of 50,265 ng/L, followed by its homologue 2-NAP (10%), both of which arose from sources including road emission, smoking and cooking. Multiple logistic regression showed that seven of the ten major PAH metabolites were correlated with increased COPD risk: including 1-NAP (OR: 1.83, 95%CI: 1.25, 2.69), 2-Hydroxyfluorene (2-FLU, OR: 2.29, 95%CI: 1.42, 3.68) and 1-Hydroxyphenanthrene (1-PHE, OR: 2.79, 95%CI: 1.85, 4.21), when compared to the lowest tertile after adjusted for covariates. Total exposure burden per PAH congener sub-group demonstrated persistent positive correlation with COPD for ∑PHE (OR: 1.80, 95%CI: 1.34, 2.43) and ∑FLU (OR: 2.74, 95%CI: 1.77, 4.23) after adjusted for covariates. To address the contribution of PAH exposure as mixture towards COPD, weighted quantile sum (WQS) regression analyses revealed that 1-NAP, 9-Hydroxyfluorene (9-FLU), 3-Hydroxyfluorene (3-FLU) and 1-PHE were among the top contributors in the associations with COPD. Our results demonstrate the contemporary yet ongoing exposure burden of PAH exposure for over a decade, particularly towards NAPs and FLUs that contribute significantly to COPD risk, calling for more timely environmental regulation.
Collapse
Affiliation(s)
- Kun Peng
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhao Li
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tian-Rui Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Jia Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Wen-Jing Wang
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ping Zhan
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wen-Cong Yao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hui Zhao
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| | - Zhu-Xia Tan
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
21
|
Brunette MF, Halenar MJ, Edwards KC, Taylor KA, Emond JA, Tanski SE, Woloshin S, Paulin LM, Hyland A, Lauten K, Mahoney M, Blanco C, Borek N, DaSilva LC, Gardner LD, Kimmel HL, Sargent JD. Association between tobacco product use and asthma among US adults from the Population Assessment of Tobacco and Health (PATH) Study waves 2-4. BMJ Open Respir Res 2023; 10:e001187. [PMID: 36750276 PMCID: PMC9906250 DOI: 10.1136/bmjresp-2021-001187] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 12/08/2022] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Research on cigarettes and adult asthma offers mixed findings, perhaps due to overlap with chronic obstructive pulmonary disease (COPD) and inadequate adjustment for other smoke exposures. Associations between other tobacco products, including e-cigarettes, and asthma are also understudied. RESEARCH QUESTION Using Population Assessment of Tobacco and Health Study waves 2-4 (2014/2015-2016/2017) data, we assessed the relation between tobacco product use and asthma in persons unlikely to have COPD. STUDY DESIGN AND METHODS Prospective study of 10 267 adults aged 18-39 years without COPD diagnoses. Past-month tobacco use at wave 2 was modelled first as combustible versus non-combustible use and second as specific product categories (former, cigarettes, e-cigarettes, cigars, hookah, smokeless tobacco). Outcomes included lifetime asthma prevalence at wave 2, incidence (waves 3 and 4) and Asthma Control Test score (lower=worse). Multivariable regressions adjusted for predictors of asthma, including other smoke exposures: cigarette pack-years, secondhand smoke and marijuana use. Sensitivity analyses examined findings when persons >39 years and those with both COPD and asthma were added, and when smoke exposure adjustments were removed. RESULTS No product, including cigarettes and e-cigarettes, was associated with prevalence or incidence of asthma. Among people with asthma at wave 2, combustible tobacco (beta=-0.86, 95% CI (-1.32 to -0.39)) and cigarettes (beta=-1.14, 95% CI (-1.66 to -0.62)) were associated with worse asthma control. No tobacco product was associated with asthma control over time. In sensitivity analyses, tobacco use became associated with incident asthma as adults >39 years and those with asthma+COPD were added, and as adjustments for other smoke exposures were omitted. INTERPRETATION Although cigarette use was associated with worse asthma control, there were no longitudinal associations between combustible tobacco or e-cigarette use and new onset or worsening asthma in these preliminary analyses. Research on tobacco and asthma should exclude COPD and adjust for smoking history and other smoke exposures.
Collapse
Affiliation(s)
- Mary F Brunette
- Research Division, Department of Psychiatry, Dartmouth Health, Lebanon, New Hampshire, USA
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
- The Dartmouth Institute for Health Policy and Clinical Practice, Dartmouth College, Lebanon, New Hampshire, USA
| | - Michael J Halenar
- Behavorial Health and Health Policy Practice, Westat, Rockville, Maryland, USA
| | - Kathryn C Edwards
- Behavorial Health and Health Policy Practice, Westat, Rockville, Maryland, USA
| | - Kristie A Taylor
- Behavorial Health and Health Policy Practice, Westat, Rockville, Maryland, USA
| | - Jennifer A Emond
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
| | - Susanne E Tanski
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
| | - Steven Woloshin
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
- The Dartmouth Institute for Health Policy and Clinical Practice, Dartmouth College, Lebanon, New Hampshire, USA
| | - Laura M Paulin
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
| | - Andrew Hyland
- Department of Health and Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kristen Lauten
- Behavorial Health and Health Policy Practice, Westat, Rockville, Maryland, USA
| | - Martin Mahoney
- Department of Health and Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Carlos Blanco
- Division of Epidemiology, Services and Prevention Research (DESPR), National Institute on Drug Abuse, Bethesda, Maryland, USA
| | - Nicolette Borek
- Office of Science, Center for Tobacco Products, Silver Spring, Maryland, USA
| | | | - Lisa D Gardner
- Office of Science, Center for Tobacco Products, Silver Spring, Maryland, USA
| | - Heather L Kimmel
- Division of Epidemiology, Services and Prevention Research (DESPR), National Institute on Drug Abuse, Bethesda, Maryland, USA
| | - James D Sargent
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
- The C Everette Koop Institute, Dartmouth College, Lebanon, New Hampshire, USA
| |
Collapse
|
22
|
Fanta CH. Advances in Evaluation and Treatment of Severe Asthma (Part One). Med Clin North Am 2022; 106:971-986. [PMID: 36280340 DOI: 10.1016/j.mcna.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
As many as 15% to 20% of patients with asthma have incompletely or poorly controlled asthma despite treatment with inhaled corticosteroids and long-acting beta-agonist bronchodilators. They are vulnerable to burdensome symptoms, limitations to their exercise capacity, and asthma attacks that can be frightening and potentially life-threatening. This article outlines a systematic approach to their evaluation, attempting to identify remediable factors that are making their asthma more severe than most other persons with asthma. This approach includes an emphasis on ensuring the correct diagnosis, minimizing exposures to stimuli that worsen airway inflammation, alleviating modifiable comorbidities such as chronic rhinosinusitis and gastroesophageal reflux, and supporting regular medication adherence and effective technique for administering inhaled medications. A basic diagnostic laboratory work-up is recommended, to be modified and amplified according to individual patient needs.
Collapse
Affiliation(s)
- Christopher H Fanta
- Partners Asthma Center, Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital, Harvard Medical School, PBB - Clinics 3, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Kumar K, Gupta PP, Verma AK, Yadav R. Assessment of prevalence and characteristics of asthma-COPD overlap among patients with chronic airflow obstruction. Monaldi Arch Chest Dis 2022; 93. [PMID: 36062505 DOI: 10.4081/monaldi.2022.2323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/07/2022] [Indexed: 11/23/2022] Open
Abstract
Given the paucity of research on asthma-chronic obstructive pulmonary disease (COPD) overlap (ACO) and the high prevalence of co-morbidities and healthcare utilization associated with it, the current study looked at the prevalence of ACO and its clinico-radiological phenotype in patients with chronic airflow obstruction. The study was conducted at a tertiary care hospital in North India. Patients over 40 with COPD or asthma were screened for inclusion in the ACO, asthma, and COPD groups. The ACO and COPD groups were further investigated. The clinical characteristics, lung functions, health-related quality of life, and radiological features of both groups were investigated and compared. ACO was discovered in 16.3% of patients with chronic airflow obstruction (asthma and COPD). The most commonly observed symptoms at presentation in the evaluated ACO patients (n=77) were shortness of breath, wheezing, cough, and expectoration (mean age at presentation: 57.9; mean duration of illness: 8.62 years). Exacerbation rates in ACO patients were significantly higher than in COPD patients (p<0.001). The ACO group had a significantly greater mean change in FEV1 post-bronchodilator in millilitres (ml) and percentage (379.61 ml and 37.72%) than the COPD group (p<0.001). The proportion of patients with emphysema was lower in the ACO group than in the COPD group (p<0.001). The ACO and COPD groups did not differ significantly in major airway wall thickness (p=0.3), but the COPD group had a significantly higher proportion of patients with vascular attenuation and distortion (p<0.001). Patients with COPD had a higher degree of hyperinflation, according to high resolution computed tomography (HRCT) indices. This study found that patients with ACO have a distinct phenotype in terms of clinical presentation and HRCT features. More research on the radiological features of ACO is required to identify the anatomical abnormalities involved in the disease's pathogenesis and to validate the radiological features of ACO.
Collapse
|
24
|
Tanabe N, Rhee CK, Park HY, Suzuki M. Editorial: The heterogeneity in COPD phenotypes. Front Med (Lausanne) 2022; 9:982121. [PMID: 36045916 PMCID: PMC9421364 DOI: 10.3389/fmed.2022.982121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/29/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- *Correspondence: Naoya Tanabe
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Masaru Suzuki
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
25
|
Harries TH, Gilworth G, Corrigan CJ, Murphy P, Hart N, Thomas M, White PT. Withdrawal of inhaled corticosteroids from patients with COPD with mild or moderate airflow limitation in primary care: a feasibility randomised trial. BMJ Open Respir Res 2022; 9:9/1/e001311. [PMID: 36041773 PMCID: PMC9438092 DOI: 10.1136/bmjresp-2022-001311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background Inhaled corticosteroids (ICS) are frequently prescribed outside guidelines to patients with chronic obstructive pulmonary disease (COPD) with mild/moderate airflow limitation and low exacerbation risk. This primary care trial explored the feasibility of identifying patients with mild/moderate COPD taking ICS, and the acceptability of ICS withdrawal. Methods Open feasibility trial. Outcome measures included prevalence of suitable participants, feasibility of their identification, their willingness-to-accept open randomisation to ICS withdrawal or continuation over 6 months follow-up. Results 392 (13%) of 2967 patients with COPD from 20 practices (209 618 population) identified as eligible for ICS withdrawal by electronic search algorithm. After individual patient record review, 243 (62%) were excluded because of: severe airflow limitation (65, 17%); one or more severe or two or more moderate COPD exacerbations in the previous year (86, 22%); asthma (15, 4%); and severe comorbidities (77, 20%). After exclusion, 149 patients with COPD were invited to participate and 61 agreed to randomisation. At clinical assessment, 10 patients exhibited undocumented airflow reversibility (forced expiratory volume in 1 s (FEV1) reversibility >12% and >200 mL); 2 had suffered two or more undocumented, moderate exacerbations in the previous year; 7 had severe airflow limitation; and 2 had normal spirometry. Finally, 40 were randomised. One patient died and one was lost to follow-up. 18 (45%) of the 38 (10 withdrawal and 8 usual care) exhibited previously undocumented FEV1 variability suggestive of asthma, supported in the withdrawal group by significant associations with elevated fractional exhaled nitric oxide (p=0.04), elevated symptom score (p=0.04), poorer quality of life (p=0.04) and atopic status (p=0.01). Conclusions Identifying primary care patients with mild/moderate COPD suitable for ICS withdrawal is feasible but requires real-time verification because of unreliable recording of exacerbations and lung function. Suitable patients accepted randomisation to ICS withdrawal or continuation for the purposes of future studies. Follow-up compliance was high. Nearly 50% of participants with a diagnosis of mild/moderate COPD demonstrated previously undocumented FEV1 variability during follow-up, mandating monitoring for at least 6 months following withdrawal to exclude undiagnosed asthma.
Collapse
Affiliation(s)
- Timothy H Harries
- School of Population Health and Environmental Sciences, King's College London, London, UK
| | - Gill Gilworth
- School of Population Health and Environmental Sciences, King's College London, London, UK
| | | | - Patrick Murphy
- Lane Fox Respiratory Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Nicholas Hart
- Lane Fox Respiratory Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Mike Thomas
- PCPS, University of Southampton, Southampton, UK
| | - Patrick T White
- School of Population Health and Environmental Sciences, King's College London, London, UK
| |
Collapse
|
26
|
Adrish M, Anand MP, Hanania NA. Phenotypes of Asthma-Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:645-655. [PMID: 35965051 DOI: 10.1016/j.iac.2022.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asthma and chronic obstructive pulmonary disease are considered unique diseases with distinct characteristics. Asthma-chronic obstructive pulmonary disease overlap is a disorder in which the clinical characteristics of asthma and chronic obstructive pulmonary disease coexist. Asthma-chronic obstructive pulmonary disease overlap is a heterogenous condition; patients can have varied clinical presentations. There are significant gender variations among different phenotypes overlap. Age of symptom onset is another important consideration. Severity of symptoms, spirometry findings, smoking history, and type of airway inflammation varies between the different phenotypes. Understanding disease pathophysiology and establishing phenotypic models will improve a precision approach.
Collapse
Affiliation(s)
- Muhammad Adrish
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Ben Taub Hospital, 1504 Taub Loop, Houston, TX 77030, USA.
| | - Mahesh P Anand
- Department of Respiratory Medicine, JSS Medical College, JSSAHER, Mysore, KA, India
| | - Nicola A Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Ben Taub Hospital, 1504 Taub Loop, Houston, TX 77030, USA
| |
Collapse
|
27
|
Saferali A, Hersh CP. Genetic Determinants in Airways Obstructive Diseases: The Case of Asthma Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:559-573. [PMID: 35965045 PMCID: PMC9379112 DOI: 10.1016/j.iac.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Genome-wide association studies (GWAS) of asthma and chronic obstructive pulmonary disease (COPD) with ever-increasing sample sizes have found multiple genetic loci associated with either disease. However, there are few intersecting loci between asthma and COPD. GWAS specifically focused on asthma-COPD overlap (ACO) have been limited by smaller sample sizes and the lack of a consistent definition of ACO that has also hampered clinical and epidemiologic studies. Other genomic techniques, such as gene expression profiling, are feasible with smaller sample sizes. Genetic analyses of objective measures of airway reactivity and allergy/T2 inflammation biomarkers in COPD studies may be another strategy to overcome limitations in ACO definitions.
Collapse
Affiliation(s)
- Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
28
|
Chen X, Jaiswal A, Costliow Z, Herbst P, Creasey EA, Oshiro-Rapley N, Daly MJ, Carey KL, Graham DB, Xavier RJ. pH sensing controls tissue inflammation by modulating cellular metabolism and endo-lysosomal function of immune cells. Nat Immunol 2022; 23:1063-1075. [PMID: 35668320 PMCID: PMC9720675 DOI: 10.1038/s41590-022-01231-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/26/2022] [Indexed: 02/08/2023]
Abstract
Extracellular acidification occurs in inflamed tissue and the tumor microenvironment; however, a systematic study on how pH sensing contributes to tissue homeostasis is lacking. In the present study, we examine cell type-specific roles of the pH sensor G protein-coupled receptor 65 (GPR65) and its inflammatory disease-associated Ile231Leu-coding variant in inflammation control. GPR65 Ile231Leu knock-in mice are highly susceptible to both bacterial infection-induced and T cell-driven colitis. Mechanistically, GPR65 Ile231Leu elicits a cytokine imbalance through impaired helper type 17 T cell (TH17 cell) and TH22 cell differentiation and interleukin (IL)-22 production in association with altered cellular metabolism controlled through the cAMP-CREB-DGAT1 axis. In dendritic cells, GPR65 Ile231Leu elevates IL-12 and IL-23 release at acidic pH and alters endo-lysosomal fusion and degradation capacity, resulting in enhanced antigen presentation. The present study highlights GPR65 Ile231Leu as a multistep risk factor in intestinal inflammation and illuminates a mechanism by which pH sensing controls inflammatory circuits and tissue homeostasis.
Collapse
Affiliation(s)
- Xiangjun Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alok Jaiswal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Paula Herbst
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth A Creasey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Noriko Oshiro-Rapley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark J Daly
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland, Helsinki, Finland
| | | | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
29
|
Clinical Assessment and Utility of Biomarkers in Asthma-Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:631-643. [DOI: 10.1016/j.iac.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
30
|
Svenningsen S, Kirby M. Imaging in Asthma-Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:601-614. [DOI: 10.1016/j.iac.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Ni H, Aye SZ, Naing C. Magnesium sulfate for acute exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2022; 5:CD013506. [PMID: 35616126 PMCID: PMC9134202 DOI: 10.1002/14651858.cd013506.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic and progressive disease, often punctuated by recurrent flare-ups or exacerbations. Magnesium sulfate, having a bronchodilatory effect, may have a potential role as an adjunct treatment in COPD exacerbations. However, comprehensive evidence of its effects is required to facilitate clinical decision-making. OBJECTIVES To assess the effects of magnesium sulfate for acute exacerbations of chronic obstructive pulmonary disease in adults. SEARCH METHODS We searched the Cochrane Airways Trials Register, CENTRAL, MEDLINE, Embase, ClinicalTrials.gov, the World Health Organization (WHO) trials portal, EU Clinical Trials Register and Iranian Registry of Clinical Trials. We also searched the proceedings of major respiratory conferences and reference lists of included studies up to 2 August 2021. SELECTION CRITERIA We included single- or double-blind parallel-group randomised controlled trials (RCTs) assessing magnesium sulfate in adults with COPD exacerbations. We excluded cross-over trials. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. Two review authors independently selected trials for inclusion, extracted data and assessed risk of bias. The primary outcomes were: hospital admissions (from the emergency room); need for non-invasive ventilation (NIV), assisted ventilation or admission to intensive-care unit (ICU); and serious adverse events. Secondary outcomes were: length of hospital stay, mortality, adverse events, dyspnoea score, lung function and blood gas measurements. We assessed confidence in the evidence using GRADE methodology. For missing data, we contacted the study investigators. MAIN RESULTS We identified 11 RCTs (10 double-blind and 1 single-blind) with a total 762 participants. The mean age of participants ranged from 62 to 76 years. Trials were single- or two-centre trials conducted in Iran, New Zealand, Nepal, Turkey, the UK, Tunisia and the USA between 2004 and 2018. We judged studies to be at low or unclear risk of bias for most of the domains. Three studies were at high risk for blinding and other biases. Intravenous magnesium sulfate versus placebo Seven studies (24 to 77 participants) were included. Fewer people may require hospital admission with magnesium infusion compared to placebo (odds ratio (OR) 0.45, 95% CI 0.23 to 0.88; number needed to treat for an additional beneficial outcome (NNTB) = 7; 3 studies, 170 participants; low-certainty evidence). Intravenous magnesium may result in little to no difference in the requirement for non-invasive ventilation (OR 0.74, 95% CI 0.31 to 1.75; very low-certainty evidence). There were no reported cases of endotracheal intubation (2 studies, 107 participants) or serious adverse events (1 study, 77 participants) in either group. Included studies did not report intensive care unit (ICU) admission or deaths. Magnesium infusion may reduce the length of hospital stay by a mean difference (MD) of 2.7 days (95% CI 4.73 days to 0.66 days; 2 studies, 54 participants; low-certainty evidence) and improve dyspnoea score by a standardised mean difference of -1.40 (95% CI -1.83 to -0.96; 2 studies, 101 participants; low-certainty evidence). We were uncertain about the effect of magnesium infusion on improving lung function or oxygen saturation. For all adverse events, the Peto OR was 0.14 (95% CI 0.02 to 1.00; 102 participants); however, the event rate was too low to reach a robust conclusion. Nebulised magnesium sulfate versus placebo Three studies (20 to 172 participants) were included. Magnesium inhalation may have little to no impact on hospital admission (OR 0.77, 95% CI 0.21 to 2.82; very low-certainty evidence) or need for ventilatory support (NIV or mechanical ventilation) (OR 0.33, 95% CI 0.01 to 8.20; very low-certainty evidence). It may result in fewer ICU admissions compared to placebo (OR 0.39, 95% CI 0.15 to 1.00; very low-certainty evidence) and improvement in dyspnoea (MD -14.37, 95% CI -26.00 to -2.74; 1 study, 20 participants; very low-certainty evidence). There were no serious adverse events reported in either group. There was one reported death in the placebo arm in one trial, but the number of participants was too small for a conclusion. There was limited evidence about the effect of magnesium inhalation on length of hospital stay, lung function outcomes or oxygen saturation. Included studies did not report adverse events. Magnesium sulfate versus ipratropium bromide A single study with 124 participants assessed nebulised magnesium sulfate plus intravenous magnesium infusion versus nebulised ipratropium plus intravenous normal saline. There was little to no difference between these groups in terms of hospital admission (OR 1.62, 95% CI 0.78 to 3.37), endotracheal intubation (OR 1.69, 95% CI 0.61 to 4.71) and length of hospital stay (MD 1.10 days, 95% CI -0.22 to 2.42), all with very low-certainty evidence. There were no data available for non-invasive ventilation, ICU admission and serious adverse events. Adverse events were not reported. AUTHORS' CONCLUSIONS: Intravenous magnesium sulfate may be associated with fewer hospital admissions, reduced length of hospital stay and improved dyspnoea scores compared to placebo. There is no evidence of a difference between magnesium infusion and placebo for NIV, lung function, oxygen saturation or adverse events. We found no evidence for ICU admission, endotracheal intubation, serious adverse events or mortality. For nebulised magnesium sulfate, we are unable to draw conclusions about its effects in COPD exacerbations for most of the outcomes. Studies reported possibly lower ICU admissions and a lesser degree of dyspnoea with magnesium inhalation compared to placebo; however, larger studies are required to yield a more precise estimate for these outcomes. Similarly, we could not identify any robust evidence for magnesium sulfate compared to ipratropium bromide. Future well-designed multicentre trials with larger samples are required, including subgroups according to severity of exacerbations and COPD phenotypes.
Collapse
Affiliation(s)
- Han Ni
- Department of Medicine, Newcastle University Medicine Malaysia, Johor, Malaysia
| | - Swe Zin Aye
- Department of Paediatrics and Child Health, Quest International University Perak, Ipoh, Malaysia
| | - Cho Naing
- Division of Tropical Health and Medicine, James Cook University, Queensland, Australia
| |
Collapse
|
32
|
Chen YC, Chang YP, Huang KT, Hsu PY, Hsiao CC, Lin MC. Unraveling the Pathogenesis of Asthma and Chronic Obstructive Pulmonary Disease Overlap: Focusing on Epigenetic Mechanisms. Cells 2022; 11:cells11111728. [PMID: 35681424 PMCID: PMC9179497 DOI: 10.3390/cells11111728] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/05/2022] [Accepted: 05/21/2022] [Indexed: 12/10/2022] Open
Abstract
Asthma and COPD overlap (ACO) is characterized by patients presenting with persistent airflow limitation and features of both asthma and COPD. It is associated with a higher frequency and severity of exacerbations, a faster lung function decline, and a higher healthcare cost. Systemic inflammation in COPD and asthma is driven by type 1 T helper (Th1) and Th2 immune responses, respectively, both of which may contribute to airway remodeling in ACO. ACO-related biomarkers can be classified into four categories: neutrophil-mediated inflammation, Th2 cell responses, arachidonic acid-eicosanoids pathway, and metabolites. Gene–environment interactions are key contributors to the complexity of ACO and are regulated by epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding RNAs. Thus, this review focuses on the link between epigenetics and ACO, and outlines the following: (I) inheriting epigenotypes without change with environmental stimuli, or epigenetic changes in response to long-term exposure to inhaled particles plus intermittent exposure to specific allergens; (II) epigenetic markers distinguishing ACO from COPD and asthma; (III) potential epigenetic drugs that can reverse oxidative stress, glucocorticoid insensitivity, and cell injury. Improved understanding of the epigenetic regulations holds great value to give deeper insight into the mechanisms, and clarify their implications for biomedical research in ACO.
Collapse
Affiliation(s)
- Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (Y.-C.C.); (C.-C.H.); (M.-C.L.); Tel.: +886-7-731-7123 (ext. 8199) (Y.-C.C. & M.-C.L.); +886-7-731-7123 (ext. 8979) (C.-C.H.)
| | - Yu-Ping Chang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
| | - Kuo-Tung Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
| | - Po-Yuan Hsu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
| | - Chang-Chun Hsiao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (Y.-C.C.); (C.-C.H.); (M.-C.L.); Tel.: +886-7-731-7123 (ext. 8199) (Y.-C.C. & M.-C.L.); +886-7-731-7123 (ext. 8979) (C.-C.H.)
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (Y.-P.C.); (K.-T.H.); (P.-Y.H.)
- Correspondence: (Y.-C.C.); (C.-C.H.); (M.-C.L.); Tel.: +886-7-731-7123 (ext. 8199) (Y.-C.C. & M.-C.L.); +886-7-731-7123 (ext. 8979) (C.-C.H.)
| |
Collapse
|
33
|
Understanding the Genetics of Asthma-COPD Overlap. Chest 2022; 161:1125-1126. [DOI: 10.1016/j.chest.2022.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 11/20/2022] Open
|
34
|
John C, Guyatt AL, Shrine N, Packer R, Olafsdottir TA, Liu J, Hayden LP, Chu SH, Koskela JT, Luan J, Li X, Terzikhan N, Xu H, Bartz TM, Petersen H, Leng S, Belinsky SA, Cepelis A, Hernández Cordero AI, Obeidat M, Thorleifsson G, Meyers DA, Bleecker ER, Sakoda LC, Iribarren C, Tesfaigzi Y, Gharib SA, Dupuis J, Brusselle G, Lahousse L, Ortega VE, Jonsdottir I, Sin DD, Bossé Y, van den Berge M, Nickle D, Quint JK, Sayers I, Hall IP, Langenberg C, Ripatti S, Laitinen T, Wu AC, Lasky-Su J, Bakke P, Gulsvik A, Hersh CP, Hayward C, Langhammer A, Brumpton B, Stefansson K, Cho MH, Wain LV, Tobin MD. Genetic Associations and Architecture of Asthma-COPD Overlap. Chest 2022; 161:1155-1166. [PMID: 35104449 PMCID: PMC9131047 DOI: 10.1016/j.chest.2021.12.674] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Some people have characteristics of both asthma and COPD (asthma-COPD overlap), and evidence suggests they experience worse outcomes than those with either condition alone. RESEARCH QUESTION What is the genetic architecture of asthma-COPD overlap, and do the determinants of risk for asthma-COPD overlap differ from those for COPD or asthma? STUDY DESIGN AND METHODS We conducted a genome-wide association study in 8,068 asthma-COPD overlap case subjects and 40,360 control subjects without asthma or COPD of European ancestry in UK Biobank (stage 1). We followed up promising signals (P < 5 × 10-6) that remained associated in analyses comparing (1) asthma-COPD overlap vs asthma-only control subjects, and (2) asthma-COPD overlap vs COPD-only control subjects. These variants were analyzed in 12 independent cohorts (stage 2). RESULTS We selected 31 independent variants for further investigation in stage 2, and discovered eight novel signals (P < 5 × 10-8) for asthma-COPD overlap (meta-analysis of stage 1 and 2 studies). These signals suggest a spectrum of shared genetic influences, some predominantly influencing asthma (FAM105A, GLB1, PHB, TSLP), others predominantly influencing fixed airflow obstruction (IL17RD, C5orf56, HLA-DQB1). One intergenic signal on chromosome 5 had not been previously associated with asthma, COPD, or lung function. Subgroup analyses suggested that associations at these eight signals were not driven by smoking or age at asthma diagnosis, and in phenome-wide scans, eosinophil counts, atopy, and asthma traits were prominent. INTERPRETATION We identified eight signals for asthma-COPD overlap, which may represent loci that predispose to type 2 inflammation, and serious long-term consequences of asthma.
Collapse
Affiliation(s)
- Catherine John
- Department of Health Sciences, University of Leicester, Leicester, England.
| | - Anna L Guyatt
- Department of Health Sciences, University of Leicester, Leicester, England
| | - Nick Shrine
- Department of Health Sciences, University of Leicester, Leicester, England
| | - Richard Packer
- Department of Health Sciences, University of Leicester, Leicester, England
| | | | - Jiangyuan Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lystra P Hayden
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Su H Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Jukka T Koskela
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, England
| | - Xingnan Li
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ
| | - Natalie Terzikhan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine and Department of Biostatistics, University of Washington, Seattle, WA
| | - Hans Petersen
- Lovelace Respiratory Research Institute, Albuquerque, NM
| | - Shuguang Leng
- Division of Epidemiology, Biostatistics, and Preventive Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM
| | | | - Aivaras Cepelis
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Levanger, Norway
| | | | - Ma'en Obeidat
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Gudmar Thorleifsson
- deCODE Genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Deborah A Meyers
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ
| | - Eugene R Bleecker
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA
| | | | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology and UW Medicine Sleep Center, Medicine, University of Washington, Seattle, WA
| | - Josée Dupuis
- Cardiovascular Health Research Unit, Department of Medicine and Department of Biostatistics, University of Washington, Seattle, WA
| | - Guy Brusselle
- Department of Biostatistics, Boston University School of Public Health, Boston, MA; Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lies Lahousse
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Victor E Ortega
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Don D Sin
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec, QC, Canada
| | - Maarten van den Berge
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, and GRIAC Research Institute, Groningen, the Netherlands
| | - David Nickle
- Global Health, University of Washington, Seattle, WA; Gossamer Bio, San Diego, CA
| | - Jennifer K Quint
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ian Sayers
- Division of Respiratory Medicine and NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England; Biodiscovery Institute, University of Nottingham, Nottingham, England
| | - Ian P Hall
- Division of Respiratory Medicine and NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, England
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, MA
| | - Tarja Laitinen
- Division of Medicine, Department of Pulmonary Diseases, Turku University Hospital, Turku, Finland; Department of Pulmonary Diseases and Clinical Allergology, University of Turku, Turku, Finland
| | - Ann C Wu
- Center for Healthcare Research in Pediatrics (CHeRP) and PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Arnulf Langhammer
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Levanger, Norway
| | - Ben Brumpton
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Clinic of Thoracic and Occupational Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, England; Leicester NIHR Biomedical Research Centre, Leicester, England
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, England; Leicester NIHR Biomedical Research Centre, Leicester, England
| |
Collapse
|
35
|
Asthma-COPD Overlap Syndrome: Recent Insights and Unanswered Questions. J Pers Med 2022; 12:jpm12050708. [PMID: 35629128 PMCID: PMC9146831 DOI: 10.3390/jpm12050708] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 01/22/2023] Open
Abstract
The term asthma-COPD overlap (ACO) has been used to identify a heterogeneous condition in which patients present with airflow limitation that is not completely reversible and clinical and inflammatory features of both asthma and chronic obstructive pulmonary disease (COPD). ACO diagnosis may be difficult in clinical practice, while controversy still exists regarding its definition, pathophysiology, and impact. Patients with ACO experience a greater disease burden compared to patients with asthma or COPD alone, but in contrast they show better response to inhaled corticosteroid treatment than other COPD phenotypes. Current management recommendations focus on defining specific and measurable treatable clinical traits, according to disease phenotypes and underlying biological mechanisms for every single patient. In this publication, we review the current knowledge on definition, pathophysiology, clinical characteristics, and management options of ACO.
Collapse
|
36
|
|
37
|
Putcha N, Woo H, McCormack MC, Fawzy A, Romero K, Davis MF, Wise RA, Diette GB, Koehler K, Matsui EC, Hansel NN. Home Dust Allergen Exposure Is Associated with Outcomes among Sensitized Individuals with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2022; 205:412-420. [PMID: 34752729 PMCID: PMC8886951 DOI: 10.1164/rccm.202103-0583oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Environmental exposures have been associated with adverse outcomes in chronic obstructive pulmonary disease (COPD). Approximately one-third of individuals with COPD have allergic sensitization, but it is unknown whether exposure to allergens in the home is associated with outcomes. Objectives: To determine the prevalence and associations of allergen sensitization with exposure to common indoor allergens with symptoms and exacerbation risk in COPD. Methods: Allergen sensitization to five common indoor allergens was assessed in former smokers with COPD. Home settled dust was assessed for presence of corresponding allergens. Sensitization and exposure status was determined and associations evaluated in adjusted models with longitudinal outcomes including symptoms, lung function, and exacerbations. Interactions were assessed between sensitization/exposure status and lung function. Measurements and Main Results: One hundred eighty-three individuals studied were on average 67.3 years of age (SD, 8.22) with average FEV1 of 53.2% (SD, 17.6%). Seventy-seven percent of participants were exposed to at least one tested allergen, and 17% had sensitization with corresponding allergen exposure. After adjustment, sensitization with exposure was associated with lower lung function (β, -8.29; 95% confidence interval [CI], -14.80 to -1.77), higher St. George's Respiratory Questionnaire Total Score (β, 6.71; 95% CI, 0.17 to 13.25), and higher exacerbation risk (odds ratio, 2.31; 95% CI, 1.11 to 4.79). Associations appeared to be more pronounced among individuals with lower lung function. Conclusions: Allergen exposures are common in COPD and associated with adverse outcomes among those with concomitant allergen sensitization. This study establishes allergens as an important home exposure that potentially could be addressed with comprehensive home environmental modification strategies to improve COPD outcomes.
Collapse
Affiliation(s)
- Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Han Woo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Meredith C. McCormack
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ashraf Fawzy
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Karina Romero
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Meghan F. Davis
- Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and
| | - Robert A. Wise
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gregory B. Diette
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kirsten Koehler
- Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and
| | | | - Nadia N. Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
38
|
GPR65 (TDAG8) inhibits intestinal inflammation and colitis-associated colorectal cancer development in experimental mouse models. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166288. [PMID: 34628032 PMCID: PMC8629932 DOI: 10.1016/j.bbadis.2021.166288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023]
Abstract
GPR65 (TDAG8) is a proton-sensing G protein-coupled receptor predominantly expressed in immune cells. Genome-wide association studies (GWAS) have identified GPR65 gene polymorphisms as an emerging risk factor for the development of inflammatory bowel disease (IBD). Patients with IBD have an elevated risk of developing colorectal cancer when compared to the general population. To study the role of GPR65 in intestinal inflammation and colitis-associated colorectal cancer (CAC), colitis and CAC were induced in GPR65 knockout (KO) and wild-type (WT) mice using dextran sulfate sodium (DSS) and azoxymethane (AOM)/DSS, respectively. Disease severity parameters such as fecal score, colon shortening, histopathology, and mesenteric lymph node enlargement were aggravated in GPR65 KO mice compared to WT mice treated with DSS. Elevated leukocyte infiltration and fibrosis were observed in the inflamed colon of GPR65 KO when compared to WT mice which may represent a cellular mechanism for the observed exacerbation of intestinal inflammation. In line with high expression of GPR65 in infiltrated leukocytes, GPR65 gene expression was increased in inflamed intestinal tissue samples of IBD patients compared to normal intestinal tissues. Moreover, colitis-associated colorectal cancer development was higher in GPR65 KO mice than WT mice when treated with AOM/DSS. Altogether, our data demonstrate that GPR65 suppresses intestinal inflammation and colitis-associated tumor development in murine colitis and CAC models, suggesting potentiation of GPR65 with agonists may have an anti-inflammatory therapeutic effect in IBD and reduce the risk of developing colitis-associated colorectal cancer.
Collapse
|
39
|
Jo YS, Hwang YI, Yoo KH, Lee MG, Jung KS, Shin KC, Yoon HK, Kim DK, Lee SY, Rhee CK. Racial Differences in Prevalence and Clinical Characteristics of Asthma-Chronic Obstructive Pulmonary Disease Overlap. Front Med (Lausanne) 2021; 8:780438. [PMID: 34881272 PMCID: PMC8645561 DOI: 10.3389/fmed.2021.780438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Background: This study examined the differences in the prevalence and clinical features of asthma–chronic obstructive pulmonary disease (COPD) overlap (ACO) with identical diagnostic criteria by race and ethnicity in two nationwide cohorts of COPD. Methods: We used data from the Korean COPD Subgroup Study (KOCOSS) and phase I of the US Genetic Epidemiology of COPD (COPDGene) study. We defined ACO by satisfying bronchodilator response (BDR) >15% and 400 ml and/or blood eosinophil count ≥300/μl. Results: The prevalences of ACO according to ethnicity were non-Hispanic white (NHW), 21.4%; African American (AA), 17.4%; and Asian, 23.8%. Asian patients with ACO were older, predominantly male, with fewer symptoms, more severe airflow limitation, and fewer comorbidities than NHW and AA patients. During 1-year follow-up, exacerbations occurred in 28.2, 22.0, and 48.4% of NHW, AA, and Asian patients with ACO, respectively. Compared to patients with non-ACO from the same racial group, the risk for exacerbation was significantly higher in NHW and Asian patients with ACO [adjusted incident rate ratio (aIRR), 1.17; 95% CI, 1.01–1.36, and aIRR, 1.37; 95% CI, 1.09–1.71 for NHW and Asian patients with ACO, respectively]. Inhaled corticosteroid (ICS) reduced the risk for future exacerbation in total patients with ACO but the effect was not significant in each racial group. Conclusions: The prevalence of ACO was similar in the two cohorts using the same diagnostic criteria. The risk for future exacerbation was significantly higher in ACO, and the use of ICS reduced the risk for exacerbation in total patients with ACO.
Collapse
Affiliation(s)
- Yong Suk Jo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Yong Il Hwang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Kwang Ha Yoo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Myung Goo Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, South Korea
| | - Ki Suck Jung
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Kyeong-Cheol Shin
- Regional Center for Respiratory Disease, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu, South Korea
| | - Hyoung Kyu Yoon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Deog Kyeom Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Yeub Lee
- Division of Pulmonary and Allergy Medicine, Department of Internal Medicine, Korea University Anam Hospital, Korea University, Seoul, South Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
40
|
Zhang C, Zhang M, Wang Y, Su X, Lei T, Yu H, Liu J. Diagnostic value of fractional exhaled nitric oxide in differentiating the asthma-COPD overlap from COPD: a systematic review and meta-analysis. Expert Rev Respir Med 2021; 16:679-687. [PMID: 34821171 DOI: 10.1080/17476348.2022.2011221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Patients with asthma-COPD overlap (ACO) account for 15-20% of chronic obstructive pulmonary disease (COPD), and the incidence increases with age. Patients with ACO have worse outcomes without proper treatment than those with COPD alone. However, the current diagnostic criteria of ACO are mainly based on symptom features and lack of objective indicators. Recently, several studies have demonstrated that fractional exhaled nitric oxide (FeNO) was higher in ACO than in COPD alone. Thus, this study aims to determine the diagnostic value of FeNO in differentiating ACO from COPD and assisting clinical decision-making. METHODS We conducted searches in the databases including PubMed, Web of Science, Cochrane Library, and Embase to extract original studies. RESULTS In all, 10 studies involving 1335 participants were included in this meta-analysis. FeNO level was significantly higher in ACO patients than in patients with COPD alone (WMD = 11.15ppb, 95%CI = 9.01‒13.28; I2 = 18.0%, p = 0.000). The sensitivity and specificity of FeNO in distinguishing ACO from COPD were both 0.71, and the area under the receiver-operating characteristic curve (AUC) was 0.76, indicating that FeNO has moderate diagnostic accuracy in differentiating ACO from COPD. CONCLUSION FeNO as an inflammatory biomarker is effective in differentiating ACO from COPD and assisting in clinical decision-making.
Collapse
Affiliation(s)
- Chuchu Zhang
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Meng Zhang
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yalei Wang
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiaojie Su
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Ting Lei
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Haichuan Yu
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China.,The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jian Liu
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lan Zhou, Gansu Province, China
| |
Collapse
|
41
|
Lu D, Chen L, Fan C, Zeng W, Fan H, Wu X, Yu H. The Value of Impulse Oscillometric Parameters and Quantitative HRCT Parameters in Differentiating Asthma-COPD Overlap from COPD. Int J Chron Obstruct Pulmon Dis 2021; 16:2883-2894. [PMID: 34703222 PMCID: PMC8541739 DOI: 10.2147/copd.s331853] [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: 07/30/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the value of impulse oscillometry (IOS) and quantitative HRCT parameters for differentiating asthma-COPD overlap (ACO) in COPD patients. Patients and Methods We enrolled 44 controls and 66 COPD patients, divided into the pure COPD group (n=40) and the ACO group (n=26). Spearman correlation analysis was utilized to assess the relationship between the quantitative HRCT and IOS parameters. A binary logistic regression analysis was performed to analyze the associations between the different variables and the risk of ACO. Receiver operating characteristic (ROC) curves were employed to identify the optimal cutoff and assess the diagnostic value of relative volume change -856 HU to -950 HU (RVC-856 to -950), decrease in the resistance from 5 Hz to 20 Hz (R5-R20) and their combination in predicting ACO. Bootstrapping validation was used to evaluate the internal validation. The concordance index (C-index) and calibration plot were calculated to assess the discrimination and calibration of the prediction model. Results Binary logistic regression analysis indicated that RVC-856 to -950 and the IOS parameters (R5-R20, R5, X5) were independently correlated with a higher risk of developing ACO after adjusting for age, sex, body mass index (BMI), history of smoking, exacerbation and atopy or allergic rhinitis. A correlation analysis showed a good correlation between the pulmonary function parameters and RVC-856 to -950, with a weaker correlation with the % area of low attenuation (LAA%) in ACO patients. Combining RVC-856 to -950 and R5-R20 to predict ACO, the AUC was 0.909, and the optimal cutoff value was >-0.62 for RVC-856 to -950 and >0.09 for R5-R20. Good calibration and favorable discrimination were displayed with a higher C-index. Conclusion More serious small airway impairment exists in ACO patients. The combination of RVC-856 to -950 and R5-R20 could be applied to differentiate ACO from COPD patients.
Collapse
Affiliation(s)
- Dongzhu Lu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Lichang Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Chaofan Fan
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Wenyi Zeng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Huizhen Fan
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiping Wu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Huapeng Yu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
42
|
Dey S, Eapen MS, Chia C, Gaikwad AV, Wark PAB, Sohal SS. Pathogenesis, clinical features of asthma COPD overlap (ACO), and therapeutic modalities. Am J Physiol Lung Cell Mol Physiol 2021; 322:L64-L83. [PMID: 34668439 DOI: 10.1152/ajplung.00121.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Both asthma and COPD are heterogeneous diseases identified by characteristic symptoms and functional abnormalities, with airway obstruction common in both diseases. Asthma COPD overlap (ACO) does not define a single disease but is a descriptive term for clinical use that includes several overlapping clinical phenotypes of chronic airways disease with different underlying mechanisms. This literature review was initiated to describe published studies, identify gaps in knowledge, and propose future research goals regarding the disease pathology of ACO, especially the airway remodelling changes and inflammation aspects. Airway remodelling occurs in asthma and COPD, but there are differences in the structures affected and the prime anatomic site at which they occur. Reticular basement membrane thickening and cellular infiltration with eosinophils and T-helper (CD4+) lymphocytes are prominent features of asthma. Epithelial squamous metaplasia, airway wall fibrosis, emphysema, bronchoalveolar lavage (BAL) neutrophilia and (CD8+) T-cytotoxic lymphocyte infiltrations in the airway wall are features of COPD. There is no universally accepted definition of ACO, nor are there clearly defined pathological characteristics to differentiate from asthma and COPD. Understanding etiological concepts within the purview of inflammation and airway remodelling changes in ACO would allow better management of these patients.
Collapse
Affiliation(s)
- Surajit Dey
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Collin Chia
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia.,Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania, Australia
| | - Archana Vijay Gaikwad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia.,Department of Respiratory and Sleep Medicine John Hunter Hospital, New Lambton Heights, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
43
|
Odimba U, Senthilselvan A, Farrell J, Gao Z. Current Knowledge of Asthma-COPD Overlap (ACO) Genetic Risk Factors, Characteristics, and Prognosis. COPD 2021; 18:585-595. [PMID: 34555990 DOI: 10.1080/15412555.2021.1980870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Asthma-COPD overlap (ACO) is a newly identified phenotype of chronic obstructive airway diseases with shared asthma and COPD features. Patients with ACO are poorly defined, and some evidence suggests that they have worse health outcomes and greater disease burden than patients with COPD or asthma. Generally, there is no evidence-based and universal definition for ACO; several consensus documents have provided various descriptions of the phenotype. In addition, the mechanisms underlying the development of ACO are not fully understood. Whether ACO is a distinct clinical entity with its particular discrete genetic determinant different from asthma and COPD alone or an intermediate phenotype with overlapping genetic markers within asthma and COPD spectrum of obstructive airway disease remains unproven. This review summarizes the current knowledge of the genetic risk factors, characteristics, and prognosis of ACO.
Collapse
Affiliation(s)
- Ugochukwu Odimba
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | | | - Jamie Farrell
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada.,Faculty of Medicine, Health Sciences Centre (Respirology Department), Memorial University, St John's, Newfoundland and Labrador, Canada
| | - Zhiwei Gao
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
44
|
Corlateanu A, Plahotniuc A, Corlateanu O, Botnaru V, Bikov A, Mathioudakis AG, Covantev S, Siafakas N. Multidimensional indices in the assessment of chronic obstructive pulmonary disease. Respir Med 2021; 185:106519. [PMID: 34175803 DOI: 10.1016/j.rmed.2021.106519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023]
Abstract
Chronic obstructive pulmonary disease (COPD), a very common disease, is the third leading cause of death worldwide. Due to the significant heterogeneity of clinical phenotypes of COPD there is no single method suitable for predicting patients' health status and outcomes, and therefore multidimensional indices, assessing different components of the disease, were developed and are recommended for clinical practice by international guidelines. Several indices have been widely accepted: BODE and its modifications, ADO, DOSE, CODEX, COTE. They differ in their composition and aim, while they are more accurate and better validated in specific settings and populations. We review the characteristics, strengths and limitations of these indices, and we discuss their role in routine management of patients with COPD, as well as in specific clinical scenarios, such as resuscitation and ceiling of care, or decisions to offer more invasive treatments. This analysis may help clinicians to use those indexes in a more practical and appropriate way.
Collapse
Affiliation(s)
- Alexandru Corlateanu
- Department of Internal Medicine, Division of Respiratory Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Stefan cel Mare street 165, 2004, Republic of Moldova.
| | - Alexandra Plahotniuc
- Department of Internal Medicine, Division of Respiratory Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Stefan cel Mare street 165, 2004, Republic of Moldova.
| | - Olga Corlateanu
- Department of Internal Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Stefan cel Mare street 165, 2004, Republic of Moldova.
| | - Victor Botnaru
- Department of Internal Medicine, Division of Respiratory Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Stefan cel Mare street 165, 2004, Republic of Moldova.
| | - Andras Bikov
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, UK; North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Alexander G Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, UK; North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Serghei Covantev
- Department of Internal Medicine, Division of Respiratory Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Stefan cel Mare street 165, 2004, Republic of Moldova.
| | - Nikolaos Siafakas
- University General Hospital, Dept. of Thoracic Medicine, Stavrakia, Heraklion, Crete, 71110 Heraklion, Greece.
| |
Collapse
|
45
|
López-Campos JL, Carrasco Hernández L, Ruiz-Duque B, Reinoso-Arija R, Caballero-Eraso C. Step-Up and Step-Down Treatment Approaches for COPD: A Holistic View of Progressive Therapies. Int J Chron Obstruct Pulmon Dis 2021; 16:2065-2076. [PMID: 34285480 PMCID: PMC8285922 DOI: 10.2147/copd.s275943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/19/2021] [Indexed: 11/30/2022] Open
Abstract
Recent advances in inhaled drugs and a clearer definition of the disease have made the task of managing COPD more complex. Different proposals have been put forward which combine all the available treatments and the different clinical presentations in an effort to select the best therapeutic options for each clinical context. As COPD is a chronic progressive disease, the escalation of therapy has traditionally been considered the most natural way to tackle it. However, the notion of COPD as a constantly progressing disease has recently been challenged and, in specific areas, this points to the possibility of a de-escalation in treatment. In this context, the clinician requires simple, specific recommendations to guide these changes in treatment in their daily clinical practice. To accomplish this, the first step must be a correct evaluation and an accurate initial preliminary diagnosis of the patient's condition. Thereafter, the first escalation in therapy must be introduced with caution as the disease progresses, since clinical trials are not designed with clinical decision-making in mind. During this escalation, three possibilities are open to change the current treatment for a different one within the same family, to increase non-pharmacological interventions or to increase the pharmacological therapies. Beyond that point, a patient with persistent symptoms represents a complex clinical scenario which requires a specialized approach, including the evaluation of different respiratory and non-respiratory comorbidities. Unfortunately, there are few de-escalation studies available, and these are mainly observational in nature. The debate on de-escalation in pharmacological treatment, therefore, involves two main discussion points: the withdrawal of bronchodilators and the withdrawal of inhaled steroids. Altogether, the scheme for modifying treatment must be more personalized than just adding molecules, and the therapeutic response and its conditioning factors should be evaluated at each step before proceeding further.
Collapse
Affiliation(s)
- Jose Luis López-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Carrasco Hernández
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Borja Ruiz-Duque
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Rocio Reinoso-Arija
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Candelaria Caballero-Eraso
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
46
|
Association between Physical Activity and Comorbidities in Spanish People with Asthma-COPD Overlap. SUSTAINABILITY 2021. [DOI: 10.3390/su13147580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are important conditions which often coexist. Higher rates of comorbidities among people with asthma-COPD overlap (ACO) may complicate clinical management. The aim of this study was to determine the prevalence of 30 different comorbidities and to analyze associations between these comorbidities and physical activity (PA) in Spanish people with ACO. Cross-sectional data from the Spanish National Health Survey 2017 were analyzed. A total of 198 Spanish people with ACO aged 15–69 years (60.6% women) were included in this study. PA was measured with the International Physical Activity Questionnaire (IPAQ) short form. Diagnosis of chronic conditions were self-reported. Associations between PA and comorbidities were analyzed using multivariable logistic regression models. The most prevalent comorbidities were chronic allergy (58.1%), chronic lumbar pain (42.4%), chronic cervical pain (38.4%), hypertension (33.3%) and arthrosis (31.8%). A PA level lower than 600 MET·min/week was significantly associated with urinary incontinence (OR = 3.499, 95% CI = 1.369–8.944) and osteoporosis (OR = 3.056, 95% CI = 1.094–8.538) in the final adjusted model. Therefore, the potential influence of PA on reducing the risk of these conditions among people with ACO should be considered, not only because of the health benefits, but also because PA can contribute to a more sustainable world.
Collapse
|
47
|
Mekov E, Nuñez A, Sin DD, Ichinose M, Rhee CK, Maselli DJ, Coté A, Suppli Ulrik C, Maltais F, Anzueto A, Miravitlles M. Update on Asthma-COPD Overlap (ACO): A Narrative Review. Int J Chron Obstruct Pulmon Dis 2021; 16:1783-1799. [PMID: 34168440 PMCID: PMC8216660 DOI: 10.2147/copd.s312560] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
Although chronic obstructive pulmonary disease (COPD) and asthma are well-characterized diseases, they can coexist in a given patient. The term asthma-COPD overlap (ACO) was introduced to describe patients that have clinical features of both diseases and may represent around 25% of COPD patients and around 20% of asthma patients. Despite the increasing interest in ACO, there are still substantial controversies regarding its definition and its position within clinical guidelines for patients with obstructive lung disease. In general, most definitions indicate that ACO patients must present with non-reversible airflow limitation, significant exposure to smoking or other noxious particles or gases, together with features of asthma. In patients with a primary diagnosis of COPD, the identification of ACO has therapeutic implication because the asthmatic component should be treated with inhaled corticosteroids and some studies suggest that the most severe patients may respond to biological agents indicated for severe asthma. This manuscript aims to summarize the current state-of-the-art of ACO. The definitions, prevalence, and clinical manifestations will be reviewed and some innovative aspects, such as genetics, epigenetics, and biomarkers will be addressed. Lastly, the management and prognosis will be outlined as well as the position of ACO in the COPD and asthma guidelines.
Collapse
Affiliation(s)
- Evgeni Mekov
- Department of Occupational Diseases, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Alexa Nuñez
- Pneumology Department, Hospital Universitari Vall d´Hebron, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Department of Medicine (Respiratory Division), University of British Columbia, Vancouver, BC, Canada
| | | | - Chin Kook Rhee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Diego Jose Maselli
- Division of Pulmonary Diseases & Critical Care, University of Texas Health, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Andréanne Coté
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Charlotte Suppli Ulrik
- Department of Pulmonary Medicine, Copenhagen University Hospital-Hvidovre, Hvidovre, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - François Maltais
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Antonio Anzueto
- Division of Pulmonary Diseases & Critical Care, University of Texas Health, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Marc Miravitlles
- Pneumology Department, Hospital Universitari Vall d´Hebron, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| |
Collapse
|
48
|
Concurrent asthma and chronic obstructive pulmonary disease in adult ED patients: A national perspective. Am J Emerg Med 2021; 49:216-225. [PMID: 34144264 DOI: 10.1016/j.ajem.2021.05.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES Emergency department (ED) visits for Asthma and Chronic Obstructive Pulmonary Disease (COPD) are common. The designation of Asthma-COPD overlap (ACO) has been used to describe patients with features of both diseases. Studies show that ACO patients may be at increased risk of poor outcomes relative to patients with either disease alone. We sought to characterize ED visits and ED-related outcomes of patients with ACO compared to patients with Asthma or COPD alone. METHODS We conducted a secondary analysis of the National Hospital Ambulatory Medical Care Survey (NHAMCS, 2005-2018) characterizing ED visits in patients ≥35 years of age with Asthma Only, COPD Only or ACO. We performed univariable and multivariable analyses adjusting for demographics to assess relevant ED outcome variables. RESULTS From 2005 to 2018, there were an estimated 8.15, 17.78 and 0.56 million ED visits for Asthma Only, COPD Only and ACO, respectively. ACO patients were younger than COPD Only patients (mean age 50.18 versus 61.79; p < 0.001). ACO patients differed in terms of sex, race and ethnicity from patients with either disease alone. When triaged, Asthma Only (adjusted odds ratio (aOR) = 11.45; 95% confidence interval (CI), 1.20-109.38) patients were more likely to require immediate care than ACO patients. Although admission rates were comparable between groups, ACO patients had a decreased mean length of ED visit compared to both Asthma Only (p < 0.001) and COPD Only (p < 0.05) patients. COPD Only patients were less likely than ACO patients to be seen in the ED in the last 72 h (aOR = 0.22; 95% CI, 0.056-0.89), receive nebulizer therapy (aOR = 0.55; 95% CI, 0.31-0.97), bronchodilators (aOR = 0.24; 95% CI, 0.12-0.48) and systemic corticosteroids (aOR = 0.18; 95% CI, 0.091-0.35). Asthma Only patients were less likely than ACO patients to undergo any imaging (aOR = 0.55; 95% CI, 0.31-0.96) and receive antibiotics (aOR = 0.46; 95% CI, 0.23-0.93). CONCLUSIONS ACO patients appear to differ demographically from patients with either disease alone in the ED. After adjustment for these demographic differences, ACO patients appear to differ with respect to several ED variables, notably respiratory therapies; however, clinical outcomes including admission and mortality rates appear to be comparable between groups.
Collapse
|
49
|
Górka K, Gross-Sondej I, Górka J, Stachura T, Polok K, Celejewska-Wójcik N, Mikrut S, Andrychiewicz A, Sładek K, Soja J. Assessment of Airway Remodeling Using Endobronchial Ultrasound in Asthma-COPD Overlap. J Asthma Allergy 2021; 14:663-674. [PMID: 34163179 PMCID: PMC8214023 DOI: 10.2147/jaa.s306421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/30/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate the structural changes of the airways using the endobronchial ultrasound (EBUS) in ACO patients compared to severe asthma and COPD patients. PATIENTS AND METHODS The study included 17 patients with ACO, 17 patients with COPD and 33 patients with severe asthma. Detailed clinical data were obtained from all participants. Basic laboratory tests were performed, including measurement of eosinophil counts in blood and serum immunoglobulin E (IgE) concentrations. All patients underwent spirometry and bronchoscopy with EBUS (a 20‑MHz ultrasound probe) to measure the total thicknesses of the bronchial walls and their particular layers in segmental bronchi of the right lower lobe. EBUS allows to distinguish five layers of the bronchial wall. Layer 1 (L1) and layer 2 (L2) were analyzed separately, while the outer layers (layers 3-5 [L3-5]) that correspond to cartilage were assessed together. RESULTS In patients with ACO the thicknesses of the L1 and L2 layers, which are mainly responsible for remodeling, were significantly greater than in patients with COPD and significantly smaller than in patients with severe asthma (median L1= 0.17 mm vs 0.16 mm vs 0.18 mm, p<0.001; median L2= 0.18 mm vs 0.17 mm vs 0.20 mm, p<0.001, respectively). The thicknesses of the total bronchial walls (L1+L2+L3-5) and L3-5 were significantly smaller in ACO and COPD patients compared to asthma patients (median L1+L2+L3-5= 1.2 mm vs 1.14 mm vs 1.31 mm, p<0.001; median L3-5= 0.85 mm vs, 0.81 mm vs 0.92 mm, p=0.001, respectively). CONCLUSION The process of structural changes in the airways assessed by EBUS is more advanced in individuals with ACO compared to patients with COPD, and less pronounced compared to patients with severe asthma. It seems that EBUS may provide useful information about differences in airway remodeling between ACO, COPD and severe asthma.
Collapse
Affiliation(s)
- Karolina Górka
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Iwona Gross-Sondej
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jacek Górka
- Centre for Intensive Care and Perioperative Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz Stachura
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Kamil Polok
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- Centre for Intensive Care and Perioperative Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Natalia Celejewska-Wójcik
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Sławomir Mikrut
- Faculty of Mining Surveying and Environmental Engineering, AGH University of Science and Technology, Kraków, Poland
| | | | - Krzysztof Sładek
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jerzy Soja
- Department of Pulmonology and Allergology, University Hospital, Kraków, Poland
- 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
50
|
Asthma/obstructive pulmonary disease overlap: update on definition, biomarkers, and therapeutics. Curr Opin Allergy Clin Immunol 2021; 20:43-47. [PMID: 31688150 DOI: 10.1097/aci.0000000000000596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Asthma/chronic obstructive pulmonary disease overlap (ACO) continues to be a poorly understood condition. This review discusses newly proposed criteria and potential biomarkers in ACO, to aid in diagnosis and research studies, and prudent therapeutic approaches. RECENT FINDINGS A global expert panel proposed an operational definition consisting of major and minor criteria as a step toward defining ACO. Serum periostin and YKL-40 may serve as biomarkers for ACO. Clinically, a reasonable therapeutic approach to ACO is the early addition of a long-acting β-agonist (LABA) and/or a long-acting muscarinic antagonist (LAMA) to an inhaled corticosteroid (ICS). SUMMARY Both the proposed criteria and the described biomarkers for ACO can help guide clinicians in identifying this condition as well as aid researchers in designing much needed future studies. In the meantime, clinicians can treat potential ACO patients using the above approach, until therapeutic studies in clearly defined ACO patients are performed.
Collapse
|